1
|
Fritz C, Feinberg D, Radhakrishnan A, Klatt K, Chan ER, Rock P, Burack R, Parameswaran R. B Cell Activating Factor Induces Drug Resistance in Hairy Cell Leukemia Variant. Biomedicines 2025; 13:890. [PMID: 40299479 PMCID: PMC12025152 DOI: 10.3390/biomedicines13040890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Background: Chemoresistance is an existing challenge faced in the treatment of the hairy cell leukemia variant (HCL-v). Classical hairy cell leukemia (HCL-c) is very sensitive to the standard of care with purine nucleoside analogs (PNAs) cladribine (cDa) and pentostatin. However, almost half of these patients eventually become less sensitive to chemotherapy and relapse. HCL-variant (HCL-v) is a biologically distinct entity from HCL-c that is not sensitive to frontline PNA therapy, and this treatment is not recommended for these patients. To address these treatment challenges, we investigated the role of B-cell activating factor (BAFF) in promoting HCL-v cell chemoresistance. Methods: Flow cytometry and quantitative PCR were used to measure the levels of BAFF and its receptors. To determine BAFF activated pathways in HCL-c and HCL-v, the Bonna-12 HCL-c cell line or HCL-v patient-derived cancer cells were stimulated with recombinat BAFF and activation of common BAFF-activated pathways, including the nonclassical nuclear factor kappa B (NF-κB) pathway, the Extracellular Signal-Regulated Kinase (Erk) and phosphatidylinositol-3 (PI-3) kinase (PI3K)/AKT serine/threonine kinase (AKT) pathways were measured by western blotting. To test whether BAFF signaling promotes chemoresistance in HCL-v, we stimulated patient-derived HCL-v cells with BAFF and performed RNA sequencing. Lastly, to confirm the functional implications of BAFF signaling in HCL-v, we treated patient-derived HCL-v cells with exogenous BAFF before treatment with cladribine. Results: We found that HCL-v patient-derived cancer cells express receptors of BAFF at varying degrees and express relatively lower levels of membrane-bound BAFF ligand expression. BAFF stimulation of these cells resulted in substantial activation of the nonclassical NF-κB pathway, which is known to promote anti-apoptotic and pro-survival effects in B-cell cancers. Conversely, in the Bonna-12 cell line, we observed constitutive activation of the nonclassical NF-κB pathway. Through RNA sequencing, we found that BAFF upregulates a myriad of genes that are known to promote chemoresistance in various cancers, including IL1, CXCL1/2, CXCL5, CXCL8, TRAF3, and PTGS2. Lastly, we found that BAFF protects these cells from cladribine-induced cell death in vitro. Conclusions: We conclude that BAFF provides chemo-protection in HCL-v cells by activating nonclassical NF-κB signaling, which results in the upregulation of multiple pro-survival or anti-apoptotic genes. Our results highlight an important role of BAFF in HCL-v resistance to chemotherapy and suggest that the BAFF blockade may enhance the chemosensitivity to PNAs in drug-resistant HCL-v patients.
Collapse
Affiliation(s)
- Claire Fritz
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (C.F.); (D.F.); (K.K.)
| | - Daniel Feinberg
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (C.F.); (D.F.); (K.K.)
| | - Akshaya Radhakrishnan
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Kayla Klatt
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (C.F.); (D.F.); (K.K.)
| | - E. Ricky Chan
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Philip Rock
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (P.R.); (R.B.)
| | - Richard Burack
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (P.R.); (R.B.)
| | - Reshmi Parameswaran
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (C.F.); (D.F.); (K.K.)
- Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA;
- Pediatric Hematology and Oncology, The Angie Fowler Adolescent & Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children’s Hospital, Cleveland, OH 44106, USA
- The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
He Z, Starkuviene V, Keese M. The Differentiation and Regeneration Potential of ABCB5 + Mesenchymal Stem Cells: A Review and Clinical Perspectives. J Clin Med 2025; 14:660. [PMID: 39941329 PMCID: PMC11818130 DOI: 10.3390/jcm14030660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are a family of multipotent stem cells that show self-renewal under proliferation, multilineage differentiation, immunomodulation, and trophic function. Thus, these cells, such as adipose tissue-derived mesenchymal stem cells (ADSCs), bone marrow-derived MSCs (BM-MSCs), and umbilical cord-derived mesenchymal stem cells (UC-MSCs), carry great promise for novel clinical treatment options. However, the challenges associated with the isolation of MSCs and the instability of their in vitro expansion remain significant barriers to their clinical application. The plasma membrane-spanning P-glycoprotein ATP-binding cassette subfamily B member 5 positive MSCs (ABCB5+ MSCs) derived from human skin specimens offer a distinctive advantage over other MSCs. They can be easily extracted from the dermis and expanded. In culture, ABCB5+ MSCs demonstrate robust innate homeostasis and a classic trilineage differentiation. Additionally, their ability to modulate the recipients' immune system highlights their potential for allogeneic applications in regenerative medicine. In this review, we primarily discuss the differentiation potential of ABCB5+ MSCs and their perspectives in regenerative medicine.
Collapse
Affiliation(s)
- Zheng He
- BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany;
- European Center of Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Straße 13-17, 68167 Mannheim, Germany
| | - Vytaute Starkuviene
- BioQuant, Heidelberg University, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany;
- Institute of Biosciences, Vilnius University Life Sciences Center, 10257 Vilnius, Lithuania
| | - Michael Keese
- Department of Vascular Surgery, Theresienkrankenhaus, Bassermannstraße 1, 68165 Mannheim, Germany
| |
Collapse
|
3
|
Doudar NA, Khattab R, Qurany EA, Reyad HR, Mostafa N. Interleukin 1 receptor associated kinase 1 gene polymorphism association with risk of rheumatological diseases in Egyptian population. Mol Biol Rep 2025; 52:135. [PMID: 39826020 DOI: 10.1007/s11033-025-10223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND Interleukin-1 receptor-associated kinase1 (IRAK1) plays a considerable role in the inflammatory signaling pathway. The current study aimed to identify any association between (rs1059703) single nucleotide polymorphism (SNP) and vulnerability to rheumatological diseases in the pediatric and adult Egyptian population. PATIENTS AND METHODS The current study included four patient groups: adult Systemic lupus erythematosus (SLE), Rheumatoid arthritis (RA), juvenile systemic lupus erythematosus (JSLE), and juvenile idiopathic arthritis (JIA). Two healthy matched age and sex groups were included as controls. Genotypes of IRAK1 (rs1059703) SNP in patients and controls were determined using the TaqMan allelic discrimination method. RESULTS The frequency of AA homozygous genotype and allele A of IRAK1 (rs1059703) SNP is higher in adult SLE patients compared to adult healthy controls (p-value < 0.005). No similar association was detected regarding RA, JSLE, or JIA. However, JSLE patients carrying the A allele have a higher SLE International Collaborating Clinics (SLICC) damage index (SDI) SDI score and a higher stage of renal biopsy than those carrying the G allele (p-value < 0.005). CONCLUSIONS Carriers of the A allele and its homozygous genotype of rs1059703 SNP are more prone to develop SLE in adult life and to have a more severe form of the disease in children in Egypt. No significant association was detected between this SNP and RA or JIA.
Collapse
Affiliation(s)
- Noha A Doudar
- Clinical and Chemical Pathology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt.
| | - Rasha Khattab
- Clinical and Chemical Pathology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Eman Abdou Qurany
- Pediatric Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Hoda Ramadan Reyad
- Medical Biochemistry Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Noha Mostafa
- Pediatric Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Wang Q, Han J, Wei M, Miao H, Zhang M, Wu B, Chen Y, Zheng Y, Gale RP, Yin B. Multi-Walled Carbon Nanotubes Accelerate Leukaemia Development in a Mouse Model. TOXICS 2024; 12:646. [PMID: 39330574 PMCID: PMC11435454 DOI: 10.3390/toxics12090646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/24/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
Inflammation is associated with an increased risk of developing various cancers in both animals and humans, primarily solid tumors but also myeloproliferative neoplasms (MPNs), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). Multi-walled carbon nanotubes (MWCNTs), a type of carbon nanotubes (CNTs) increasingly used in medical research and other fields, are leading to a rising human exposure. Our study demonstrated that exposing mice to MWCNTs accelerated the progression of spontaneous MOL4070LTR virus-induced leukemia. Additionally, similar exposures elevated pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and induced reactive oxygen species (ROS) in a murine macrophage cell line. These effects were significantly reduced in immunodeficient mice and when mice were treated with methoxypolyethylene glycol amine (PEG)-modified MWCNTs. These findings underscore the necessity of evaluating the safety of MWCNTs, particularly for those with hematologic cancers.
Collapse
Affiliation(s)
- Qingqing Wang
- Clinical Medical Research Center, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China; (Q.W.); (J.H.); (M.W.)
| | - Jingdan Han
- Clinical Medical Research Center, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China; (Q.W.); (J.H.); (M.W.)
| | - Mujia Wei
- Clinical Medical Research Center, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China; (Q.W.); (J.H.); (M.W.)
| | - Huikai Miao
- Department of Laboratory Medicine, Jiangnan University Medical Center, Wuxi 214002, China; (H.M.); (M.Z.); (B.W.); (Y.C.)
| | - Min Zhang
- Department of Laboratory Medicine, Jiangnan University Medical Center, Wuxi 214002, China; (H.M.); (M.Z.); (B.W.); (Y.C.)
| | - Biao Wu
- Department of Laboratory Medicine, Jiangnan University Medical Center, Wuxi 214002, China; (H.M.); (M.Z.); (B.W.); (Y.C.)
| | - Yao Chen
- Department of Laboratory Medicine, Jiangnan University Medical Center, Wuxi 214002, China; (H.M.); (M.Z.); (B.W.); (Y.C.)
| | - Yanwen Zheng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China;
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial College London, London SW7 2AZ, UK;
| | - Bin Yin
- Clinical Medical Research Center, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China; (Q.W.); (J.H.); (M.W.)
- Department of Laboratory Medicine, Jiangnan University Medical Center, Wuxi 214002, China; (H.M.); (M.Z.); (B.W.); (Y.C.)
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China;
| |
Collapse
|
5
|
Luo Y, Su B, Hung V, Luo Y, Shi Y, Wang G, de Graaf D, Dinarello CA, Spaner DE. IL-1 receptor antagonism reveals a yin-yang relationship between NFκB and interferon signaling in chronic lymphocytic leukemia. Proc Natl Acad Sci U S A 2024; 121:e2405644121. [PMID: 39121163 PMCID: PMC11331101 DOI: 10.1073/pnas.2405644121] [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: 03/26/2024] [Accepted: 07/10/2024] [Indexed: 08/11/2024] Open
Abstract
Nuclear factor kappa B (NFκB) is a pathogenic factor in chronic lymphocytic leukemia (CLL) that is not addressed specifically by current therapies. NFκB is activated by inflammatory factors that stimulate toll-like receptors (TLRs) and receptors for interleukin-1 (IL-1) family members. IL-1 is considered a master regulator of inflammation, and IL-1 receptor signaling is inhibited by the IL-1 receptor antagonist anakinra. These considerations suggested that anakinra might have a role in the treatment of CLL. Consistent with this idea, anakinra inhibited spontaneous and TLR7-mediated activation of the canonical NFκB pathway in CLL cells in vitro. However, CLL cells exhibited only weak signaling responses to IL-1 itself, and anakinra was found to inhibit NFκB along with oxidative stress in an IL-1 receptor-independent manner. Anakinra was then administered with minimal toxicity to 11 previously untreated CLL patients in a phase I dose-escalation trial (NCT04691765). A stereotyped clinical response was observed in all patients. Anakinra lowered blood lymphocytes and lymph node sizes within the first month that were associated with downregulation of NFκB and oxidative stress in the leukemia cells. However, inhibition of NFκB was accompanied by upregulation of type 1 interferon (IFN) signaling, c-MYC-regulated genes and proteins, and loss of the initial clinical response. Anakinra increased IFN signaling and survival of CLL cells in vitro that were, respectively, phenocopied by mitochondrial antioxidants and reversed by IFN receptor blocking antibodies. These observations suggest that anakinra has activity in CLL and may be a useful adjunct for conventional therapies as long as compensatory IFN signaling is blocked at the same time.
Collapse
Affiliation(s)
- YuXuan Luo
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
- Department of Immunology, University of Toronto, TorontoM5S 1A8, Canada
| | - BoYang Su
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
- Department of Medical Biophysics, University of Toronto, TorontoM5G 2M9, Canada
| | - Vincent Hung
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
| | - YuHan Luo
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
- Department of Immunology, University of Toronto, TorontoM5S 1A8, Canada
| | - Yonghong Shi
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
| | - Guizhi Wang
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
| | - Dennis de Graaf
- Department of Medicine, University of Colorado Denver, Denver, CO80045
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn53127, Germany
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Denver, CO80045
- Department of Medicine, Radboud University Medical Center, Nijmegen6525 GA, The Netherlands
| | - David E. Spaner
- Biological Science Platform, Sunnybrook Research Institute, Sunnybrook hospital, TorontoM4N 3M5, Canada
- Department of Immunology, University of Toronto, TorontoM5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, TorontoM5G 2M9, Canada
- Department of Hematology, Odette Cancer Center, TorontoM4N 3M5, Canada
- Department of Medicine, University of Toronto, TorontoM5G 2C4, Canada
| |
Collapse
|
6
|
Tran D, Beeler JS, Liu J, Wiley B, Chan IC, Xin Z, Kramer MH, Batchi-Bouyou AL, Zong X, Walter MJ, Petrone GE, Chlamydas S, Ferraro F, Oh ST, Link DC, Busby B, Cao Y, Bolton KL. Plasma Proteomic Signature Predicts Myeloid Neoplasm Risk. Clin Cancer Res 2024; 30:3220-3228. [PMID: 38446993 PMCID: PMC11292192 DOI: 10.1158/1078-0432.ccr-23-3468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/10/2024] [Accepted: 03/04/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Clonal hematopoiesis (CH) is thought to be the origin of myeloid neoplasms (MN). Yet, our understanding of the mechanisms driving CH progression to MN and clinical risk prediction of MN remains limited. The human proteome reflects complex interactions between genetic and epigenetic regulation of biological systems. We hypothesized that the plasma proteome might predict MN risk and inform our understanding of the mechanisms promoting MN development. EXPERIMENTAL DESIGN We jointly characterized CH and plasma proteomic profiles of 46,237 individuals in the UK Biobank at baseline study entry. During 500,036 person-years of follow-up, 115 individuals developed MN. Cox proportional hazard regression was used to test for an association between plasma protein levels and MN risk. RESULTS We identified 115 proteins associated with MN risk, of which 30% (N = 34) were also associated with CH. These were enriched for known regulators of the innate and adaptive immune system. Plasma proteomics improved the prediction of MN risk (AUC = 0.85; P = 5×10-9) beyond clinical factors and CH (AUC = 0.80). In an independent group (N = 381,485), we used inherited polygenic risk scores (PRS) for plasma protein levels to validate the relevance of these proteins toMNdevelopment. PRS analyses suggest that most MN-associated proteins we identified are not directly causally linked toMN risk, but rather represent downstream markers of pathways regulating the progression of CH to MN. CONCLUSIONS These data highlight the role of immune cell regulation in the progression of CH to MN and the promise of leveraging multi-omic characterization of CH to improveMN risk stratification. See related commentary by Bhalgat and Taylor, p. 3095.
Collapse
Affiliation(s)
- Duc Tran
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - J. Scott Beeler
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Jie Liu
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Brian Wiley
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Irenaeus C.C. Chan
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Zilan Xin
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Michael H. Kramer
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Armel L. Batchi-Bouyou
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Xiaoyu Zong
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.
| | - Matthew J. Walter
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Giulia E.M. Petrone
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | | | - Francesca Ferraro
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Stephen T. Oh
- Division of Hematology, Department of Medicine, WUSM, St. Louis, Missouri.
| | - Daniel C. Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| | - Ben Busby
- DNAnexus, Mountain View, California.
| | - Yin Cao
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.
| | - Kelly L. Bolton
- Division of Oncology, Department of Medicine, Washington University School of Medicine (WUSM), St. Louis, Missouri.
| |
Collapse
|
7
|
Chen Y, Ni J, Wang C, Zhai X, Luo T, Li YP, Wei Y, Liu Y. The proteomic analysis uncovers the cellular responses to the African swine fever virus membrane proteins p54, p17, and pB117L. Microbes Infect 2024; 26:105348. [PMID: 38697277 DOI: 10.1016/j.micinf.2024.105348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024]
Abstract
African swine fever virus (ASFV) infection causes African swine fever (ASF), a highly contagious and fatal disease that poses severe threat to swine production. To gain insights into the host responses to ASFV, we generated recombinant adenovirus Ad5 expressing viral membrane proteins p54, p17, and pB117L individually and infected an alveolar cell line, 3D4/21, with these recombinant viruses. Then, the cell lysates were analyzed using label-free quantification proteomic analysis method. A total of 2158 differentially expressed proteins (DEPs) were identified, of which 817, 466, and 875 proteins were from Ad5-p54-, Ad5-p17-, Ad5-pB117L-infected 3D4/21 cells, respectively. Gene Ontology (GO) classification and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed distinct yet interconnecting patterns of protein interaction networks. Specifically, the Ad5-p54 virus infection enriched the DEPs primarily involved in the metabolic pathways, endocytosis, adherens junction, and SNARE interactions in vesicular transport. The Ad5-p17 virus infection enriched the DEPs in endocytosis, ubiquitin-mediated proteolysis, N-Glycan biosynthesis, and apoptosis, while the Ad5-pB117L virus infection enriched the DEPs in metabolic pathways, endocytosis, oxidative phosphorylation, and focal adhesion. In summary, these results provide a comprehensive proteinomics analysis of the cellular responses to three ASFV membrane proteins, thus facilitating our understanding of ASFV pathogenesis.
Collapse
Affiliation(s)
- Yuhong Chen
- College of Animal Science and Technology, Guangxi University, Nanning 530005, Guangxi, China; China Animal Disease Control Center (CADC), Beijing 102618, China
| | - Jianqiang Ni
- China Animal Disease Control Center (CADC), Beijing 102618, China
| | - Chuanbin Wang
- China Animal Disease Control Center (CADC), Beijing 102618, China
| | - Xinyan Zhai
- China Animal Disease Control Center (CADC), Beijing 102618, China
| | - Tingrong Luo
- College of Animal Science and Technology, Guangxi University, Nanning 530005, Guangxi, China
| | - Yi-Ping Li
- Institute of Human Virology, Department of Pathogen Biology and Biosecurity, and Key Laboratory of Tropical Disease Control of Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Youchuan Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530005, Guangxi, China.
| | - Yuliang Liu
- China Animal Disease Control Center (CADC), Beijing 102618, China.
| |
Collapse
|
8
|
Liu C, Lan Q, Cao S, Zheng F, Liang Y, Shen J, Wang Y, Ikezoe T, Xu K, Pan B. Thrombin receptor activating peptide-6 decreases acute graft-versus-host disease through activating GPR15. Leukemia 2024; 38:1390-1402. [PMID: 38459169 DOI: 10.1038/s41375-024-02212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/10/2024]
Abstract
G-protein coupled receptor 15 (GPR15) is expressed on T-cells. We previously reported knockout of GPR15 increased acute graft-versus-host disease (GvHD) in mice. In this study, we identified thrombin receptor activating peptide-6 (TRAP-6, peptide sequence: SFLLRN) as an activator of GPR15. GRP15 and β-arrestin2 were needed for TRAP-6-mediated inhibition of mixed lymphocyte reactions. TRAP-6 decreased acute GvHD in allotransplant models in mice, an effect dependent on GPR15-expression in donor T-cells. RNA-seq and protein analyses indicated TRAP-6 increased binding of β-arrestin2/TAB1 and inhibited phosphorylation of TAK1 and NF-κB-P65. GPR15 is expressed differently on CD4+ T-cells and CD8+ T-cells. TRAP-6 inhibited phosphorylation of NF-κB-P65 in CD4+ T-cells but increased granzyme B expression in CD8+ T-cells. TRAP-6 decreased acute GvHD without inhibiting graft-versus-tumor (GvT) efficacy against A20 lymphoma cells. SALLRN, a mutant of TRAP-6, preserved the anti-acute GvHD effect but avoided the adverse effects of TRAP-6. TRAP-6 and SALLRN also decreased allogeneic and xenogeneic reactions induced by human blood mononuclear cells. In conclusion, TRAP-6 activated GPR15 on T-cells and decreased acute GvHD in mice without impairing GvT efficacy.
Collapse
Affiliation(s)
- Cong Liu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Qiu Lan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Shuo Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Fei Zheng
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Yiwen Liang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Jingyi Shen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Ying Wang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Takayuki Ikezoe
- Department of Hematology, Fukushima Medical University, Fukushima, Japan.
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| | - Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
9
|
Rai S, Zhang Y, Grockowiak E, Kimmerlin Q, Hansen N, Stoll CB, Usart M, Luque Paz D, Hao-Shen H, Zhu Y, Roux J, Bader MS, Dirnhofer S, Farady CJ, Schroeder T, Méndez-Ferrer S, Skoda RC. IL-1β promotes MPN disease initiation by favoring early clonal expansion of JAK2-mutant hematopoietic stem cells. Blood Adv 2024; 8:1234-1249. [PMID: 38207211 PMCID: PMC10912850 DOI: 10.1182/bloodadvances.2023011338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
ABSTRACT JAK 2-V617F is the most frequent somatic mutation causing myeloproliferative neoplasm (MPN). JAK2-V617F can be found in healthy individuals with clonal hematopoiesis of indeterminate potential (CHIP) with a frequency much higher than the prevalence of MPNs. The factors controlling the conversion of JAK2-V617F CHIP to MPN are largely unknown. We hypothesized that interleukin-1β (IL-1β)-mediated inflammation can favor this progression. We established an experimental system using bone marrow (BM) transplantations from JAK2-V617F and GFP transgenic (VF;GFP) mice that were further crossed with IL-1β-/- or IL-1R1-/- mice. To study the role of IL-1β and its receptor on monoclonal evolution of MPN, we performed competitive BM transplantations at high dilutions with only 1 to 3 hematopoietic stem cells (HSCs) per recipient. Loss of IL-1β in JAK2-mutant HSCs reduced engraftment, restricted clonal expansion, lowered the total numbers of functional HSCs, and decreased the rate of conversion to MPN. Loss of IL-1R1 in the recipients also lowered the conversion to MPN but did not reduce the frequency of engraftment of JAK2-mutant HSCs. Wild-type (WT) recipients transplanted with VF;GFP BM that developed MPNs had elevated IL-1β levels and reduced frequencies of mesenchymal stromal cells (MSCs). Interestingly, frequencies of MSCs were also reduced in recipients that did not develop MPNs, had only marginally elevated IL-1β levels, and displayed low GFP-chimerism resembling CHIP. Anti-IL-1β antibody preserved high frequencies of MSCs in VF;GFP recipients and reduced the rate of engraftment and the conversion to MPN. Our results identify IL-1β as a potential therapeutic target for preventing the transition from JAK2-V617F CHIP to MPNs.
Collapse
Affiliation(s)
- Shivam Rai
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yang Zhang
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Elodie Grockowiak
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Quentin Kimmerlin
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nils Hansen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Cedric B. Stoll
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marc Usart
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Damien Luque Paz
- University of Angers, Nantes Université, CHU Angers, INSERM, CNRS, CRCI2NA, Angers, France
| | - Hui Hao-Shen
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Yexuan Zhu
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Julien Roux
- Department of Biomedicine, Bioinformatics core facility, University of Basel, Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michael S. Bader
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Stefan Dirnhofer
- Department of Pathology, University Hospital Basel, Basel, Switzerland
| | | | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Radek C. Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
10
|
Zhou Z, Zhou X, Jiang X, Yang B, Lu X, Fei Y, Zhao L, Chen H, Zhang L, Si X, Liang N, Wang Y, Yang D, Peng Y, Yang Y, Yao Z, He Y, Wu X, Zhang W, Wang M, Yang H, Zhang X. Single-cell profiling identifies IL1B hi macrophages associated with inflammation in PD-1 inhibitor-induced inflammatory arthritis. Nat Commun 2024; 15:2107. [PMID: 38453911 PMCID: PMC10920757 DOI: 10.1038/s41467-024-46195-x] [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: 07/08/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024] Open
Abstract
Inflammatory arthritis (IA) is a common rheumatic adverse event following immune checkpoint inhibitors treatment. The clinical disparities between IA and rheumatoid arthritis (RA) imply disease heterogeneity and distinct mechanisms, which remain elusive. Here, we profile CD45+ cells from the peripheral blood or synovial fluid (SF) of patients with PD-1-induced IA (PD-1-IA) or RA using single-cell RNA sequencing. We report the predominant expansion of IL1Bhi myeloid cells with enhanced NLRP3 inflammasome activity, in both the SF and peripheral blood of PD-1-IA, but not RA. IL1Bhi macrophages in the SF of PD-1-IA shared similar inflammatory signatures and might originate from peripheral IL1Bhi monocytes. Exhausted CD8+ T cells (Texs) significantly accumulated in the SF of patients with PD-1-IA. IL1Bhi myeloid cells communicated with CD8+ Texs possibly via the CCR1-CCL5/CCL3 and CXCL10-CXCR3 axes. Collectively, these results demonstrate different cellular and molecular pathways in PD-1-IA and RA and highlight IL1Bhi macrophages as a possible therapeutic target in PD-1-IA.
Collapse
Affiliation(s)
- Ziyue Zhou
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xiaoxiang Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Xu Jiang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- National Infrastructure for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Bo Yang
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xin Lu
- Department of Orthopedics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xiaoyan Si
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Dan Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yezi Peng
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Yiying Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Zhuoran Yao
- Department of Thoracic Oncology, Cancer Center, and Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Yangzhige He
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- National Infrastructure for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Xunyao Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, 100730, Beijing, China
| | - Huaxia Yang
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases, the Ministry of Education Key Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China.
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, 100730, Beijing, China.
| |
Collapse
|
11
|
Marzoog BA. Cytokines and Regulating Epithelial Cell Division. Curr Drug Targets 2024; 25:190-200. [PMID: 38213162 DOI: 10.2174/0113894501279979240101051345] [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: 09/27/2023] [Revised: 11/19/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Abstract
Physiologically, cytokines play an extremely important role in maintaining cellular and subcellular homeostasis, as they interact almost with every cell in the organism. Therefore, cytokines play a significantly critical role in the field of pathogenic pharmacological therapy of different types of pathologies. Cytokine is a large family containing many subfamilies and can be evaluated into groups according to their action on epithelial cell proliferation; stimulatory include transforming growth factor-α (TGF-α), Interlukine-22 (IL-22), IL-13, IL-6, IL-1RA and IL-17 and inhibitory include IL-1α, interferon type I (IFN type I), and TGF-β. The balance between stimulatory and inhibitory cytokines is essential for maintaining normal epithelial cell turnover and tissue homeostasis. Dysregulation of cytokine production can contribute to various pathological conditions, including inflammatory disorders, tissue damage, and cancer. Several cytokines have shown the ability to affect programmed cell death (apoptosis) and the capability to suppress non-purpose cell proliferation. Clinically, understanding the role of cytokines' role in epithelial tissue is crucial for evaluating a novel therapeutic target that can be of use as a new tactic in the management of carcinomas and tissue healing capacity. The review provides a comprehensive and up-to-date synthesis of current knowledge regarding the multifaceted effects of cytokines on epithelial cell proliferation, with a particular emphasis on the intestinal epithelium. Also, the paper will highlight the diverse signaling pathways activated by cytokines and their downstream consequences on epithelial cell division. It will also explore the potential therapeutic implications of targeting cytokine- epithelial cell interactions in the context of various diseases.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| |
Collapse
|
12
|
Ahmed Najar I, Sharma A, Alshammari A, Albekairi TH, Alharbi M, Ahmad Dar T, Latief Qadrie Z, Kabra A, Newton A, Kumar M. Anti-oxidant and anti-inflammatory potential of different polymer-based mesalamine delayed-release granules in TNBS-induced ulcerative colitis in wistar rats. Saudi Pharm J 2024; 32:101910. [PMID: 38111669 PMCID: PMC10727943 DOI: 10.1016/j.jsps.2023.101910] [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: 08/16/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory condition of colon characterized by severe damage to the innermost colon tissues. A number of studies described the use of medication delivery systems based on natural polymers like polysaccharides for the purpose of reaching the colon. In this research, polymer-based mesalamine delayed-release granules (DRGs) were tested for their antioxidant and anti-inflammatory efficacy against UC. Chitosan (C), pectin (P), and pectin-chitosan (PC) mesalamine (M) DRGs were prepared and characterized. Data revealed satisfactory compatibility, flow, packing properties, drug release pattern, and delayed drug release by DRGs. Wistar rats were treated with 2,4,6-trinitrobenzenesulfonic acid (TNBS) (100 mg/kg) via rectal administration. Mesalamine and mesalamine DRGs (50 mg/kg) were administered orally separately for 14 days. Biomarkers of oxidative stress, inflammation, hematological tests, colon profile, and histopathology were performed. The findings demonstrated the good efficacy of the polysaccharides in delivering mesalamine to colon. Mesalamine and mesalamine DRGs based on various polymers showed significant antioxidant and anti-inflammatory effects in rats with UC. Mesalamine granules significantly attenuated colon lipid peroxidation, nitrites, myeloperoxidase activity, and interleukin-1β levels, and improved anti-oxidants (GSH, SOD). Data showed upregulation of Nrf2 activity by mesalamine granules with CM-DRGs showing maximum effect. Mesalamine and different polymer-based mesalamine DRGs significantly attenuated TNBS-induced decline in body weight, ulcer severity, and colon damage. CM-DRGs showed the most pronounced ameliorative effect on colon and hematology parameters via anti-oxidant and anti-inflammatory activities. Chitosan can be used as a carrier for oral colon delivery of mesalamine in DRG formulation for enhanced therapeutic efficacy in UC.
Collapse
Affiliation(s)
- Imtiyaz Ahmed Najar
- Department of Pharmacology, Lovely Professional University, Jalandhar, Punjab, India
| | - Archana Sharma
- Department of Pharmacology, Swift School of Pharmacy, Rajpura, Punjab, India
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Thamer H. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Taief Ahmad Dar
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu & Kashmir, India
| | - Zulfkar Latief Qadrie
- Department of Clinical Pharmacology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, Jammu & Kashmir, India
| | - Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - A.M.J Newton
- Lowy Cancer Research Centre, Prince of Wales Clinical School, University of New South Wales, Randwick, Australia
| | - Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
13
|
Chavez JS, Rabe JL, Niño KE, Wells HH, Gessner RL, Mills TS, Hernandez G, Pietras EM. PU.1 is required to restrain myelopoiesis during chronic inflammatory stress. Front Cell Dev Biol 2023; 11:1204160. [PMID: 37497478 PMCID: PMC10368259 DOI: 10.3389/fcell.2023.1204160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic inflammation is a common feature of aging and numerous diseases such as diabetes, obesity, and autoimmune syndromes and has been linked to the development of hematological malignancy. Blood-forming hematopoietic stem cells (HSC) can contribute to these diseases via the production of tissue-damaging myeloid cells and/or the acquisition of mutations in epigenetic and transcriptional regulators that initiate evolution toward leukemogenesis. We previously showed that the myeloid "master regulator" transcription factor PU.1 is robustly induced in HSC by pro-inflammatory cytokines such as interleukin (IL)-1β and limits their proliferative activity. Here, we used a PU.1-deficient mouse model to investigate the broader role of PU.1 in regulating hematopoietic activity in response to chronic inflammatory challenges. We found that PU.1 is critical in restraining inflammatory myelopoiesis via suppression of cell cycle and self-renewal gene programs in myeloid-biased multipotent progenitor (MPP) cells. Our data show that while PU.1 functions as a key driver of myeloid differentiation, it plays an equally critical role in tailoring hematopoietic responses to inflammatory stimuli while limiting expansion and self-renewal gene expression in MPPs. These data identify PU.1 as a key regulator of "emergency" myelopoiesis relevant to inflammatory disease and leukemogenesis.
Collapse
Affiliation(s)
- James S. Chavez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer L. Rabe
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Katia E. Niño
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Harrison H. Wells
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel L. Gessner
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Taylor S. Mills
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Giovanny Hernandez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eric M. Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
14
|
Chen L, Wang Z, Wu J, Yao Q, Peng J, Zhang C, Chen H, Li Y, Jiang Z, Liu Y, Shi C. Released dsDNA-triggered inflammasomes serve as intestinal radioprotective targets. Clin Transl Immunology 2023; 12:e1452. [PMID: 37333051 PMCID: PMC10276537 DOI: 10.1002/cti2.1452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 05/05/2023] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Objectives Intestinal mucositis is the major side effect during abdominal or pelvic radiotherapy, but the underlying immunogen remains to be further characterised and few radioprotective agents are available. This study investigated the role of dsDNA-triggered inflammasomes in intestinal mucositis during radiotherapy. Methods Pro-inflammatory cytokines were detected by ELISA. Radiation-induced intestinal injury in mice was analyzed by means of survival curves, body weight, HE staining of intestines, and intestinal barrier integrity. Western blot, immunofluorescence staining, co-immunoprecipitation assay and flow cytometry were used to investigate the regulatory role of dsDNA on inflammasomes. Results Here, we show that a high level of IL-1β and IL-18 is associated with diarrhoea in colorectal cancer (CRC) patients during radiotherapy, which accounts for intestinal radiotoxicity. Subsequently, we found that the dose-dependently released dsDNA from the intestinal epithelial cells (IECs) serves as the potential immunogenic molecule for radiation-induced intestinal mucositis. Our results further indicate that the released dsDNA transfers into the macrophages in an HMGB1/RAGE-dependent manner and then triggers absent in melanoma 2 (AIM2) inflammasome activation and the IL-1β and IL-18 secretion. Finally, we show that the FDA-approved disulfiram (DSF), a newly identified inflammasome inhibitor, could mitigate intestinal radiotoxicity by controlling inflammasome. Conclusion These findings indicate that the extracellular self-dsDNA released from the irradiated IECs is a potential immunogen to stimulate immune cells and trigger the subsequent intestinal mucositis, while blunting the dsDNA-triggered inflammasome in macrophages may represent an exciting therapeutic strategy for side effects control during abdominal radiotherapy.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
- Shigatse Branch, Xinqiao Hospital, Army 953 HospitalArmy Medical UniversityShigatseChina
| | - Ziwen Wang
- Department of CardiologyGeriatric Cardiovascular Disease Research and Treatment Center, 252 Hospital of PLABaodingChina
| | - Jie Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Quan Yao
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Center, School of Medicine, Sichuan Cancer Hospital & InstituteUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jingjing Peng
- Department of OncologyWestern Theater General HospitalChengduChina
| | - Chi Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Hongdan Chen
- Breast and Thyroid Surgical Department, Chongqing General HospitalUniversity of Chinese Academy of SciencesChongqingChina
| | - Yingjie Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Zhongyong Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Yunsheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| | - Chunmeng Shi
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Rocket Force MedicineArmy Medical UniversityChongqingChina
| |
Collapse
|
15
|
Blijlevens NMA, de Mooij CEM. Mucositis and Infection in Hematology Patients. Int J Mol Sci 2023; 24:ijms24119592. [PMID: 37298545 DOI: 10.3390/ijms24119592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Survival in patients with hematological malignancies has improved over the years, both due to major developments in anticancer treatment, as well as in supportive care. Nevertheless, important and debilitating complications of intensive treatment regimens still frequently occur, including mucositis, fever and bloodstream infections. Exploring potential interacting mechanisms and directed therapies to counteract mucosal barrier injury is of the utmost importance if we are to continue to improve care for this increasingly growing patient population. In this perspective, I highlight recent advances in our understanding of the relation of mucositis and infection.
Collapse
Affiliation(s)
- Nicole M A Blijlevens
- Department of Haematology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Charlotte E M de Mooij
- Department of Haematology, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
16
|
Frankle L, Riley A, Tomor R, Lee H, Jarzembak K, Benedict O, Sternbach S, Shelestak J, McDonough J, Clements R. Changes to Astrocyte-associated Protein Expression at Different Timepoints of Cuprizone Treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537627. [PMID: 37131767 PMCID: PMC10153238 DOI: 10.1101/2023.04.20.537627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Glial cells, including astrocytes, microglia, and oligodendrocytes, are brain cells that support and dynamically interact with neurons and each other. These intercellular dynamics undergo changes during stress and disease states. In response to most forms of stress, astrocytes will undergo some variation of activation, meaning upregulation in certain proteins expressed and secreted and either upregulations or downregulations to various constitutive and normal functions. While types of activation are many and contingent on the particular disturbance that triggers these changes, there are two main overarching categories that have been delineated thus far: A1 and A2. Named in the convention of microglial activation subtypes, and with the acknowledgement that the types are not completely distinct or completely comprehensive, the A1 subtype is generically associated with toxic and pro-inflammatory factors, and the A2 phenotype is broadly associated with anti-inflammatory and neurogenic factors. The present study served to measure and document dynamic changes in these subtypes at multiple timepoints using an established experimental model of cuprizone toxic demyelination. The authors found increases in proteins associated with both cell types at different timepoints, with protein increases in the A1 marker C3d and the A2 marker Emp1 in the cortex at one week and protein increases in Emp1 in the corpus callosum at three days and four weeks. There were also increases in Emp1 staining specifically colocalized with astrocyte staining in the corpus callosum at the same timepoints as the protein increases, and in the cortex weeks later at four weeks. C3d colocalization with astrocytes also increased most at four weeks. This indicates simultaneous increases of both types of activation as well as the likely existence of astrocytes expressing both markers. The authors also found the increase in two A1 associated proteins (TNF alpha and C3d) did not show a linear relationship in line with findings from other research and indicating a more complex relationship between cuprizone toxicity and astrocyte activation. The increases in TNF alpha and IFN gamma did not occur at timepoints preceding increases in C3d and Emp1, showing that other factors also precipitate the subtypes associated (A1 for C3d and A2 for Emp1). These findings add to the body of research showing the specific early timepoints at which A1 and A2 markers are most increased during the course of cuprizone treatment, including the fact that these increases can be non-linear in the case of Emp1. This provides additional information on optimal times for targeted interventions during the cuprizone model.
Collapse
Affiliation(s)
- Lana Frankle
- Kent State University Biological Sciences Department
| | - Amanda Riley
- Kent State University Biological Sciences Department
| | - Riely Tomor
- Kent State University Biological Sciences Department
| | - Hannah Lee
- Kent State University Biological Sciences Department
| | | | | | | | | | | | | |
Collapse
|
17
|
The Role of NLRP3, a Star of Excellence in Myeloproliferative Neoplasms. Int J Mol Sci 2023; 24:ijms24054860. [PMID: 36902299 PMCID: PMC10003372 DOI: 10.3390/ijms24054860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) is the most widely investigated inflammasome member whose overactivation can be a driver of several carcinomas. It is activated in response to different signals and plays an important role in metabolic disorders and inflammatory and autoimmune diseases. NLRP3 belongs to the pattern recognition receptors (PRRs) family, expressed in numerous immune cells, and it plays its primary function in myeloid cells. NLRP3 has a crucial role in myeloproliferative neoplasms (MPNs), considered to be the diseases best studied in the inflammasome context. The investigation of the NLRP3 inflammasome complex is a new horizon to explore, and inhibiting IL-1β or NLRP3 could be a helpful cancer-related therapeutic strategy to improve the existing protocols.
Collapse
|
18
|
Lai W, Liao J, Li X, Liang P, He L, Huang K, Liang X, Wang Y. Characterization of the microenvironment in different immune-metabolism subtypes of cervical cancer with prognostic significance. Front Genet 2023; 14:1067666. [PMID: 36816023 PMCID: PMC9935837 DOI: 10.3389/fgene.2023.1067666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Immune cell infiltration and metabolic reprogramming may have great impact on the tumorigenesis and progression of malignancies. The interaction between these two factors in cervical cancer remains to be clarified. Here we constructed a gene set containing immune and metabolism related genes and we applied this gene set to molecular subtyping of cervical cancer. Methods: Bulk sequencing and single-cell sequencing data were downloaded from the Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database respectively. Immune and metabolism related genes were collected from Immport and Kyoto encyclopedia of genes and genomes (KEGG) database respectively. Unsupervised consensus clustering was performed to identify the molecular subtypes. Cibersort was applied to evaluate the immune cells infiltration status. Differential expression analysis and Gene set enrichment analysis (GSEA) were performed to characterize the molecular pattern of different subtypes. Multivariate Cox regression analysis was used for prognosis prediction model construction and receiver operating characteristic (ROC) curve was used for performance evaluation. The hub genes in the model were verified in single-cell sequencing dataset and clinical specimens. In vitro experiments were performed to validate the findings in our research. Results: Three subtypes were identified with prognostic implications. C1 subgroup was in an immunosuppressive state with activation of mitochondrial cytochrome P450 metabolism, C2 had poor immune cells infiltration and was characterized by tRNA anabolism, and the C3 subgroup was in an inflammatory state with activation of aromatic amino acid synthesis. The area under the ROC curve of the constructed model was 0.8, which showed better performance than clinical features. IMPDH1 was found to be significantly upregulated in tumor tissue and it was demonstrated that IMPDH1 could be a novel therapeutic target in vitro. Discussion: In summary, our findings suggested novel molecular subtypes of cervical cancer with distinct immunometabolic profiles and uncovered a novel therapeutic target.
Collapse
Affiliation(s)
- Wujiang Lai
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jinrong Liao
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Li
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peili Liang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liqing He
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Keke Huang
- Department of Obstetrics, Shunde Hospital, The First People’s Hospital of Shunde, Southern Medical University, Foshan, Guangdong, China,*Correspondence: Keke Huang, ; Xiaomei Liang, ; Yifeng Wang,
| | - Xiaomei Liang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,*Correspondence: Keke Huang, ; Xiaomei Liang, ; Yifeng Wang,
| | - Yifeng Wang
- Obstetrics and Gynecology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China,*Correspondence: Keke Huang, ; Xiaomei Liang, ; Yifeng Wang,
| |
Collapse
|
19
|
Mohammed AI, Celentano A, Paolini R, Low JT, McCullough MJ, O' Reilly LA, Cirillo N. Characterization of a novel dual murine model of chemotherapy-induced oral and intestinal mucositis. Sci Rep 2023; 13:1396. [PMID: 36697446 PMCID: PMC9876945 DOI: 10.1038/s41598-023-28486-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Oral and intestinal mucositis are debilitating inflammatory diseases observed in cancer patients undergoing chemo-radiotherapy. These are devastating clinical conditions which often lead to treatment disruption affecting underlying malignancy management. Although alimentary tract mucositis involves the entire gastrointestinal tract, oral and intestinal mucositis are often studied independently utilizing distinct organ-specific pre-clinical models. This approach has however hindered the development of potentially effective whole-patient treatment strategies. We now characterize a murine model of alimentary tract mucositis using 5-Fluorouracil (5-FU). Mice were given 5-FU intravenously (50 mg/kg) or saline every 48 h for 2 weeks. Post initial injection, mice were monitored clinically for weight loss and diarrhea. The incidence and extent of oral mucositis was assessed macroscopically. Microscopical and histomorphometric analyses of the tongue and intestinal tissues were conducted at 3 interim time points during the experimental period. Repeated 5-FU treatment caused severe oral and intestinal atrophy, including morphological damage, accompanied by body weight loss and mild to moderate diarrhea in up to 77.8% of mice. Oral mucositis was clinically evident throughout the observation period in 88.98% of mice. Toluidine blue staining of the tongue revealed that the ulcer size peaked at day-14. In summary, we have developed a model reproducing the clinical and histologic features of both oral and intestinal mucositis, which may represent a useful in vivo pre-clinical model for the study of chemotherapy-induced alimentary tract mucositis and the development of preventative therapies.
Collapse
Affiliation(s)
- Ali I Mohammed
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia. .,College of Dentistry, The University of Tikrit, Tikrit, Iraq.
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Jun T Low
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Michael J McCullough
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia
| | - Lorraine A O' Reilly
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3000, Australia
| | - Nicola Cirillo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC, 3053, Australia.
| |
Collapse
|
20
|
Niebergall-Roth E, Frank NY, Ganss C, Frank MH, Kluth MA. Skin-Derived ABCB5 + Mesenchymal Stem Cells for High-Medical-Need Inflammatory Diseases: From Discovery to Entering Clinical Routine. Int J Mol Sci 2022; 24:66. [PMID: 36613507 PMCID: PMC9820160 DOI: 10.3390/ijms24010066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
The ATP-binding cassette superfamily member ABCB5 identifies a subset of skin-resident mesenchymal stem cells (MSCs) that exhibit potent immunomodulatory and wound healing-promoting capacities along with superior homing ability. The ABCB5+ MSCs can be easily accessed from discarded skin samples, expanded, and delivered as a highly homogenous medicinal product with standardized potency. A range of preclinical studies has suggested therapeutic efficacy of ABCB5+ MSCs in a variety of currently uncurable skin and non-skin inflammatory diseases, which has been substantiated thus far by distinct clinical trials in chronic skin wounds or recessive dystrophic epidermolysis bullosa. Therefore, skin-derived ABCB5+ MSCs have the potential to provide a breakthrough at the forefront of MSC-based therapies striving to fulfill current unmet medical needs. The most recent milestones in this regard are the approval of a phase III pivotal trial of ABCB5+ MSCs for treatment of recessive dystrophic and junctional epidermolysis bullosa by the US Food and Drug Administration, and national market access of ABCB5+ MSCs (AMESANAR®) for therapy-refractory chronic venous ulcers under the national hospital exemption pathway in Germany.
Collapse
Affiliation(s)
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Ganss
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| | - Markus H. Frank
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Mark A. Kluth
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Cassanello G, Pasquale R, Barcellini W, Fattizzo B. Novel Therapies for Unmet Clinical Needs in Myelodysplastic Syndromes. Cancers (Basel) 2022; 14:4941. [PMID: 36230864 PMCID: PMC9562187 DOI: 10.3390/cancers14194941] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/01/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a very heterogeneous disease, with extremely variable clinical features and outcomes. Current management relies on risk stratification based on IPSS and IPSS-R, which categorizes patients into low (LR-) and high-risk (HR-) MDS. Therapeutic strategies in LR-MDS patients mainly consist of erythropoiesis stimulating agents (ESAs), transfusion support, and luspatercept or lenalidomide for selected patients. Current unmet needs include the limited options available after treatment failure, and the consequent transfusion burden with several hospital admissions and poor quality of life. Therapeutic approaches in HR-MDS patients are aimed at changing the natural course of the disease and hypometylating agents (HMA) are the first choice. The only potentially curative treatment is allogeneic stem cell transplant (allo-HCT), restricted to a minority of young and fit candidates. Patients unfit for or those that relapse after the abovementioned options harbor an adverse prognosis, with limited overall survival and frequent leukemic evolution. Recent advances in genetic mutations and intracellular pathways that are relevant for MDS pathogenesis are improving disease risk stratification and highlighting therapeutic targets addressed by novel agents. Several drugs are under evaluation for LR and HR patients, which differ by their mechanism of action, reported efficacy, and phase of development. This review analyzes the current unmet clinical needs for MDS patients and provides a critical overview of the novel agents under development in this setting.
Collapse
Affiliation(s)
- Giulio Cassanello
- Department of Oncology and Oncohematology, University of Milan, 20122 Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Raffaella Pasquale
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Wilma Barcellini
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Bruno Fattizzo
- Department of Oncology and Oncohematology, University of Milan, 20122 Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
22
|
Luciano M, Krenn PW, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia. Front Immunol 2022; 13:1000996. [PMID: 36248849 PMCID: PMC9554002 DOI: 10.3389/fimmu.2022.1000996] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous malignancy of the blood and bone marrow, characterized by clonal expansion of myeloid stem and progenitor cells and rapid disease progression. Chemotherapy has been the first-line treatment for AML for more than 30 years. Application of recent high-throughput next-generation sequencing technologies has revealed significant molecular heterogeneity to AML, which in turn has motivated efforts to develop new, targeted therapies. However, due to the high complexity of this disease, including multiple driver mutations and the coexistence of multiple competing tumorigenic clones, the successful incorporation of these new agents into clinical practice remains challenging. These continuing difficulties call for the identification of innovative therapeutic approaches that are effective for a larger cohort of AML patients. Recent studies suggest that chronic immune stimulation and aberrant cytokine signaling act as triggers for AML initiation and progression, facets of the disease which might be exploited as promising targets in AML treatment. However, despite the greater appreciation of cytokine profiles in AML, the exact functions of cytokines in AML pathogenesis are not fully understood. Therefore, unravelling the molecular basis of the complex cytokine networks in AML is a prerequisite to develop new therapeutic alternatives based on targeting cytokines and their receptors.
Collapse
Affiliation(s)
- Michela Luciano
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Peter W. Krenn
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
23
|
Wardill HR, de Mooij CEM, Da Silva Ferreira AR, Havinga H, Harmsen HJM, van der Velden WJFM, van Groningen LFJ, Tissing WJE, Blijlevens NMA. Supporting the gastrointestinal microenvironment during high-dose chemotherapy and stem cell transplantation by inhibiting IL-1 signaling with anakinra. Sci Rep 2022; 12:6803. [PMID: 35546555 PMCID: PMC9095632 DOI: 10.1038/s41598-022-10700-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 04/07/2022] [Indexed: 11/20/2022] Open
Abstract
High-dose chemotherapy causes intestinal inflammation and subsequent breakdown of the mucosal barrier, permitting translocation of enteric pathogens, clinically manifesting as fever. Antibiotics are mainstay for controlling these complications, however, they are increasingly recognized for their detrimental effects, including antimicrobial resistance and dysbiosis. Here, we show that mucosal barrier injury induced by the mucotoxic chemotherapeutic agent, high-dose melphalan (HDM), is characterized by hyper-active IL-1b/CXCL1/neutrophil signaling. Inhibition of this pathway with IL-1RA, anakinra, minimized the duration and intensity of mucosal barrier injury and accompanying clinical symptoms, including diarrhea, weight loss and fever in rats. 16S analysis of fecal microbiome demonstrated a more stable composition in rats receiving anakinra, with reduced pathogen expansion. In parallel, we report through Phase IIA investigation that anakinra is safe in stem cell transplant patients with multiple myeloma after HDM. Ramping-up anakinra (100–300 mg administered intravenously for 15 days) did not cause any adverse events or dose limiting toxicities, nor did it change time to neutrophil recovery. Our results reinforce that strengthening the mucosal barrier may be an effective supportive care strategy to mitigate local and systemic clinical consequences of HDM. We are now conducting a Phase IIB multicenter, placebo-controlled, double-blinded trial to assess clinical efficacy of anakinra (AFFECT-2). Trial registration: ClinicalTrials.gov identifier: NCT03233776.
Collapse
Affiliation(s)
- H R Wardill
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia. .,The Supportive Oncology Research Group, Precision Medicine Theme (Cancer), The South Australian Health and Medical Research Institute, Adelaide, SA, Australia. .,Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - C E M de Mooij
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A R Da Silva Ferreira
- Department of Medical Microbiology, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H Havinga
- Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H J M Harmsen
- Department of Medical Microbiology, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - L F J van Groningen
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W J E Tissing
- Department of Pediatrics, The University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Princes Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - N M A Blijlevens
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Sun X, Xia M, Liu J, Cui J, Zhang Y, Sun R, Cui X. lnc-AC145676.2.1-6-3 plays an important role in intestinal acute graft-versus-host disease through the regulation of interleukin-1β. Int J Lab Hematol 2022; 44:759-768. [PMID: 35441492 DOI: 10.1111/ijlh.13852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/26/2022] [Accepted: 04/03/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Acute graft-versus-host disease (aGVHD) is one of the major complications of allogeneic hematopoietic stem cell transplantation, and the liver, skin, and gastrointestinal tract are the main target organs. The most common type is intestinal aGVHD. Long noncoding RNAs (lncRNAs) have coregulatory functions and participate in a variety of intracellular regulatory processes. We investigated the expression of lncRNAs and their mechanisms in the development of aGVHD. METHODS The participants included 15 patients with aGVHD and 4 healthy controls (HCs). To generate profiles of abnormally expressed lncRNAs, peripheral blood mononuclear cell (PBMC) lncRNAs from four patients and four HCs were validated by high-throughput sequencing and quantitative real-time-PCR (qRT-PCR). A number of databases, including Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, miRanda, TargetScan, and Metascape, were used for bioinformatics analysis. Bioinformatics analysis indicated that overexpression of lnc-AC145676.2.1-6-3 might induce aGVHD via the interleukin (IL)-1β axis and a downstream miRNA. After the higher levels of lnc-AC145676.2.1-6-3 in other patients were confirmed by qRT-PCR, serum IL-1β, IL-6, and tumor necrosis factor-α were measured by enzyme linked immunosorbent assays. RESULTS In our study, a large number of lncRNAs were found in PBMCs of patients with intestinal aGVHD, and bioinformatics analysis showed that the upregulated lncRNA lnc-AC145676.2.1-6-3 probably affected the progression of intestinal aGVHD by regulating the hsa-miR-3064-5p/IL-1β axis. In addition, the changes in lncRNA expression levels were positively correlated with the clinical characteristics of intestinal aGVHD. CONCLUSION Our results suggest that lncRNAs in PBMCs may become new biomarkers and therapeutic targets for intestinal aGVHD.
Collapse
Affiliation(s)
- Xiaoqi Sun
- Department of Traditional Chinese Medicine, Shandong University of Traditional, Chinese Medicine, Jinan, China
| | - Mengting Xia
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiang Liu
- Department of Osteoporosis, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinwei Cui
- Jingwu Road Primary School of Jinan, Jinan, China
| | - Yanyu Zhang
- Department of Traditional Chinese Medicine, Shandong University of Traditional, Chinese Medicine, Jinan, China
| | - Runjie Sun
- Department of Traditional Chinese Medicine, Shandong University of Traditional, Chinese Medicine, Jinan, China
| | - Xing Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.,Center of Oncology and Hematology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine/Shandong Hospital of Integrated Traditional Chinese and Western Medicine, Jinan, China
| |
Collapse
|
25
|
Bazinet A, Bravo GM. New Approaches to Myelodysplastic Syndrome Treatment. Curr Treat Options Oncol 2022; 23:668-687. [PMID: 35320468 DOI: 10.1007/s11864-022-00965-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2022] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT The treatment of myelodysplastic syndromes (MDS) begins with risk stratification using a validated tool such as the International Prognostic Scoring System (IPSS) or its revised version (IPSS-R). This divides patients into lower- and higher- risk categories. Although treatment objectives in lower-risk MDS (LR-MDS) have traditionally been directed at improving cytopenias (usually anemia) as well as quality of life, recent data supports a potential role for early intervention in delaying transfusion dependency. In addition, careful individualized risk stratification incorporating clinical, cytogenetic, and mutational data might help identify patients at higher-than-expected risk for progression. Given the need for supportive care with red blood cell (RBC) transfusions leading to iron overload, iron chelation should be considered for patients with heavy transfusion requirements at risk for end-organ complications. For patients with LR-MDS and isolated anemia, no high-risk features, and endogenous erythropoietin (EPO) levels below 500 U/L, erythropoiesis-stimulating agents (ESAs) can be attempted to improve anemia. Some LR-MDS patient subgroups may also benefit from specific therapies including luspatercept (MDS with ring sideroblasts), lenalidomide (MDS with deletion 5q), or immunosuppressive therapy (hypocellular MDS). LR-MDS patients failing the above options, or those with multiple cytopenias and/or higher-risk features, can be considered for oral low-dose hypomethylating agent (HMA) therapy. Alternatively, these patients may be enrolled on a clinical trial with promising agents targeting the transforming-growth factor beta (TGF-β) pathway, the hypoxia-inducible factor (HIF) pathway, telomerase activity, inflammatory signaling, or the splicing machinery. In higher-risk MDS (HR-MDS), therapy seeks to modify the natural history of the disease and prolong survival. Eligible patients should be considered for curative allogeneic hematopoietic stem cell transplantation (aHSCT). Despite promising novel combinations, the HMAs azacitidine (AZA) or decitabine (DAC) are still the standard of care for these patients, with intensive chemotherapy-based approaches being a potential option in a small subset of patients. Individuals who fail to respond or progress after HMA experience dismal outcomes and represent a major unmet clinical need. Such patients should be treated as part of a clinical trial if possible. Experimental agents to consider include venetoclax, myeloid cell leukemia 1 (MCL-1) inhibitors, eprenetapopt, CPX-351, immunotherapies (directed towards CD47, TIM3, or CD70), interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitors, pevonedistat, seclidemstat, and eltanexor. In this review, we extensively discuss the current landscape of experimental therapies for both LR- and HR-MDS.
Collapse
Affiliation(s)
- Alexandre Bazinet
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Box 428, Houston, TX, 77030, USA
| | - Guillermo Montalban Bravo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Box 428, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Andina N, Bonadies N, Allam R. Inflammasome Activation in Myeloid Malignancies—Friend or Foe? Front Cell Dev Biol 2022; 9:825611. [PMID: 35155452 PMCID: PMC8829542 DOI: 10.3389/fcell.2021.825611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022] Open
Abstract
Myeloid malignancies including myelodysplastic syndromes, myeloproliferative neoplasms and acute myeloid leukemia are heterogeneous disorders originating from mutated hematopoietic stem and progenitor cells (HSPCs). Genetically, they are very heterogeneous and characterized by uncontrolled proliferation and/or blockage of differentiation of abnormal HSPCs. Recent studies suggest the involvement of inflammasome activation in disease initiation and clonal progression. Inflammasomes are cytosolic innate immune sensors that, upon activation, induce caspase-1 mediated processing of interleukin (IL) -1-cytokine members IL-1β and IL-18, as well as initiation of gasdermin D-dependent pyroptosis. Inflammasome activation leads to a pro-inflammatory microenvironment in the bone marrow, which drives proliferation and may induce clonal selection of mutated HSPCs. However, there are also contradictory data showing that inflammasome activation actually counteracts leukemogenesis. Overall, the beneficial or detrimental effect of inflammasome activation seems to be highly dependent on mutational, environmental, and immunological contexts and an improved understanding is fundamental to advance specific therapeutic targeting strategies. This review summarizes current knowledge about this dichotomous effect of inflammasome activation in myeloid malignancies and provides further perspectives on therapeutic targeting.
Collapse
Affiliation(s)
- Nicola Andina
- Department of Hematology and Central Hematology Laboratory, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Nicolas Bonadies
- Department of Hematology and Central Hematology Laboratory, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ramanjaneyulu Allam
- Department of Hematology and Central Hematology Laboratory, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- *Correspondence: Ramanjaneyulu Allam,
| |
Collapse
|
27
|
da Silva Ferreira AR, van der Aa SAJ, Wehkamp T, Wardill HR, Ten Klooster JP, Garssen J, Harthoorn LF, Hartog A, Harmsen HJM, Tissing WJE, van Bergenhenegouwen J. Development of a self-limiting model of methotrexate-induced mucositis reinforces butyrate as a potential therapy. Sci Rep 2021; 11:22911. [PMID: 34824316 PMCID: PMC8617074 DOI: 10.1038/s41598-021-02308-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
Gastrointestinal mucositis is a complication of anticancer treatment, with few validated in vitro systems suitable to study the complex mechanisms of mucosal injury. Therefore, we aimed to develop and characterize a chemotherapeutic-induced model of mucositis using 3D intestinal organoids. Organoids derived from mouse ileum were grown for 7 days and incubated with different concentrations of the chemotherapeutic agent methotrexate (MTX). Metabolic activity, citrulline levels and cytokine/chemokine production were measured to determine the optimal dosage and incubation time. The protective effects of folinic acid on the toxicity of MTX were investigated by pre-treating organoids with (0.0005-50 µg/mL) folinic acid. The impact of microbial-derived short-chain fatty acids was evaluated by supplementation with butyrate in the organoid model. MTX caused a dose-dependent reduction in cell metabolic activity and citrulline production that was salvaged by folinic acid treatment. Overall, MTX causes significant organoid damage, which can be reversed upon removal of MTX. The protective effect of folinic acid suggest that the organoids respond in a clinical relevant manner. By using the model for intervention, it was found that prophylactic treatment with butyrate might be a valuable strategy for prophylactic mucositis prevention.
Collapse
Affiliation(s)
- A R da Silva Ferreira
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Center Groningen, Hanzeplein 1 EB80, 9713 GZ, Groningen, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - S A J van der Aa
- Department of Pediatric Oncology, University of Groningen, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
- Danone Nutricia Research, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - T Wehkamp
- Danone Nutricia Research, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - H R Wardill
- Department of Pediatric Oncology, University of Groningen, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
- Adelaide Medical School, The University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - J P Ten Klooster
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - J Garssen
- Danone Nutricia Research, Utrecht, The Netherlands
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - L F Harthoorn
- Danone Nutricia Research, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - A Hartog
- Danone Nutricia Research, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| | - H J M Harmsen
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Center Groningen, Hanzeplein 1 EB80, 9713 GZ, Groningen, The Netherlands.
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands.
| | - W J E Tissing
- Department of Pediatric Oncology, University of Groningen, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - J van Bergenhenegouwen
- Danone Nutricia Research, Utrecht, The Netherlands
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Research Centre for Healthy and Sustainable Living, Innovative Testing in Life Sciences and Chemistry, University of Applied Sciences, Utrecht, The Netherlands
| |
Collapse
|
28
|
The Impact of NLRP3 Activation on Hematopoietic Stem Cell Transplantation. Int J Mol Sci 2021; 22:ijms222111845. [PMID: 34769275 PMCID: PMC8584591 DOI: 10.3390/ijms222111845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023] Open
Abstract
NLR family pyrin domain-containing 3 (NLRP3) is an intracellular protein that after recognizing a broad spectrum of stressors, such as microbial motifs and endogenous danger signals, promotes the activation and release of the pro-inflammatory cytokines IL-1β and IL-18, thus playing an essential role in the innate immune response. Several blood cell types, including macrophages, dendritic cells, and hematopoietic stem and progenitor cells (HSPCs), express NLRP3, where it has been implicated in various physiological and pathological processes. For example, NLRP3 participates in the development and expansion of HSPCs, and their release from bone marrow into the peripheral blood has been implicated in certain hematological disorders including various types of leukemia. In addition, accumulating evidence indicates that activation of NLRP3 plays a pivotal role in the development of transplant complications in patients receiving hematopoietic stem cell transplantation (HSCT) including graft versus host disease, severe infections, and transplant-related mortality. The majority of these complications are triggered by the severe tissue damage derived from the conditioning regimens utilized in HSCT which, in turn, activates NLRP3 and, ultimately, promotes the release of proinflammatory cytokines such as IL-1β and IL-18. Here, we summarize the implications of NLRP3 in HSCT with an emphasis on the involvement of this inflammasome component in transplant complications.
Collapse
|
29
|
IL-1 Mediates Microbiome-Induced Inflamm-Ageing of Hematopoietic Stem Cells in Mice. Blood 2021; 139:44-58. [PMID: 34525198 DOI: 10.1182/blood.2021011570] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/30/2021] [Indexed: 11/20/2022] Open
Abstract
Ageing is associated with impaired hematopoietic and immune function. This is caused in part by decreased hematopoietic stem cell (HSC) population fitness and an increased myeloid differentiation bias. The reasons for this aging-associated HSC impairment are incompletely understood. We here demonstrate that aged specific pathogen free (SPF) wild-type mice in contrast to young SPF mice produce more IL-1a/b in steady-state bone marrow (BM), with most of IL-1a/b being derived from myeloid BM cells. Further, blood of steady-state aged SPF wild-type mice contains higher levels of microbe associated molecular patterns (MAMPs), specifically TLR4 and TLR8 ligands. Also, BM myeloid cells from aged mice produce more IL-1b in vitro, and aged mice show higher and more durable IL-1a/b responses upon LPS stimulation in vivo. To test if HSC ageing is driven via IL-1a/b, we evaluated HSCs from IL-1 receptor 1 (IL-1R1) knock-out mice. Indeed, aged HSCs from IL-1R1 knock-out mice show significantly mitigated ageing-associated inflammatory signatures. Moreover, HSCs from aged IL-1R1KO and also from germ-free mice maintain unbiased lympho-myeloid hematopoietic differentiation upon transplantation, thus resembling this functionality of young HSCs. Importantly, in vivo antibiotic suppression of microbiota or pharmacologic blockade of IL-1 signaling in aged wild-type mice was similarly sufficient to reverse myeloid biased output of their HSC populations. Collectively, our data defines the microbiome-IL-1/IL-1R1 axis as a key, self-sustaining, but also therapeutically partially reversible driver of HSC inflamm-ageing.
Collapse
|
30
|
Saul-McBeth J, Dillon J, Lee A, Launder D, Kratch JM, Abutaha E, Williamson AA, Schroering AG, Michalski G, Biswas P, Conti SR, Shetty AC, McCracken C, Bruno VM, Parsai EI, Conti HR. Tissue Damage in Radiation-Induced Oral Mucositis Is Mitigated by IL-17 Receptor Signaling. Front Immunol 2021; 12:687627. [PMID: 34220843 PMCID: PMC8248500 DOI: 10.3389/fimmu.2021.687627] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 01/13/2023] Open
Abstract
Oral mucositis (OM) is a treatment-limiting adverse side effect of radiation and chemotherapy. Approximately 80% of patients undergoing radiotherapy (RT) for head and neck cancers (HNC) develop OM, representing a major unmet medical condition. Our understanding of the immunopathogenesis of OM is limited, due in part to the surprising paucity of information regarding healing mechanisms in the oral mucosa. RNAseq of oral tissue in a murine model that closely mimics human OM, showed elevated expression of IL-17 and related immune pathways in response to head and neck irradiation (HNI). Strikingly, mice lacking the IL-17 receptor (IL-17RA) exhibited markedly more severe OM. Restoration of the oral mucosa was compromised in Il17ra-/- mice and components associated with healing, including matrix metalloproteinase 3, 10 and IL-24 were diminished. IL-17 is typically associated with recruitment of neutrophils to mucosal sites following oral infections. Unexpectedly, in OM the absence of IL-17RA resulted in excessive neutrophil recruitment and immunopathology. Instead, neutrophil activation was IL-1R-driven in Il17ra-/- mice. Blockade of IL-1R and depletion of neutrophils lessened the severity of damage in these mice. Overall, we show IL-17 is protective in OM through multiple mechanisms including restoration of the damaged epithelia and control of the neutrophil response. We also present a clinically relevant murine model of human OM to improve mechanistic understanding and develop rational translational therapeutics.
Collapse
Affiliation(s)
- Jessica Saul-McBeth
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - John Dillon
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Aaron Lee
- Department of Radiation Oncology, Division of Medical Physics, The University of Toledo, Toledo, OH, United States
| | - Dylan Launder
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Jacqueline M. Kratch
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Eanas Abutaha
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | | | | | - Grace Michalski
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Priosmita Biswas
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Samuel R. Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| | - Amol C. Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Carrie McCracken
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Vincent M. Bruno
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - E. Ishmael Parsai
- Department of Radiation Oncology, Division of Medical Physics, The University of Toledo, Toledo, OH, United States
| | - Heather R. Conti
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
31
|
de Mooij CEM, van Groningen LFJ, de Haan AFJ, Biemond BJ, Bakker M, van der Velden WJFM, Blijlevens NMA. Anakinra: efficacy in the management of fever during neutropenia and mucositis in autologous stem cell transplantation (AFFECT-2)-study protocol for a multicenter randomized double-blind placebo-controlled trial. Trials 2020; 21:948. [PMID: 33225965 PMCID: PMC7681989 DOI: 10.1186/s13063-020-04847-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/28/2020] [Indexed: 02/02/2023] Open
Abstract
Background Since decades, fever and infections have been the most important complications of intensive chemotherapy and hematopoietic stem cell transplantation (HSCT) in the treatment of hematologic malignancies. Neutropenia has long been considered to be the most important risk factor for these complications. However, recent studies have shown that not neutropenia, but the development of mucositis is the most important cause of these complications. Currently, limited options for the prevention and treatment of mucositis are available, of which most are only supportive. The pro-inflammatory cytokine interleukin-1 (IL-1) plays a crucial role in the pathogenesis of mucositis. Pre-clinical studies of chemotherapy-induced mucositis have shown that recombinant human IL-1 receptor antagonist anakinra significantly ameliorated intestinal mucositis. In our pilot study AFFECT-1, we examined the safety and maximal tolerated dose of anakinra in patients with multiple myeloma, treated with high-dose melphalan (HDM) and autologous HSCT, selecting a dose of 300 mg daily for the phase IIb trial. The aim of the AFFECT-2 study is to determine the efficacy of anakinra in preventing fever during neutropenia (FN) and mucositis in this study population. Methods/design A multicenter, randomized, placebo-controlled, double-blind phase IIb trial will be conducted. Ninety patients with multiple myeloma scheduled for treatment with HDM and autologous HSCT will be included. Patients will be randomized between intravenous treatment with anakinra (300 mg) or placebo. Each group will be treated from day − 2 (day of HDM; day 0 is HSCT) up until day + 12. Outcome measures will be assessed at baseline, during admission, at discharge or day + 30, at day + 90, and + 1 year. The primary outcome will be reduction of FN. Secondary outcome measures include mucositis scores, bloodstream infections, citrulline levels, quality of life, and fatigue severity. Discussion The AFFECT-2 trial will examine the efficacy of anakinra in the management of fever during neutropenia and mucositis in patients with multiple myeloma treated with HDM and autologous HSCT. The results of this study may provide a new treatment option for these important complications. Also, this study will give us more insight in the pathophysiology of mucositis, including the role of IL-1 and the role of the microbiota in mucositis. Trial registration Clinicaltrials.gov NCT04099901. Registered on September 23, 2019. EudraCT: 2018-005046-10.
Collapse
Affiliation(s)
- Charlotte E M de Mooij
- Radboud Institute of Health Sciences, Department of Hematology, Radboud University Medical Center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands.
| | - Lenneke F J van Groningen
- Radboud Institute of Health Sciences, Department of Hematology, Radboud University Medical Center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Anton F J de Haan
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bart J Biemond
- Department of Hematology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Martijn Bakker
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Walter J F M van der Velden
- Radboud Institute of Health Sciences, Department of Hematology, Radboud University Medical Center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| | - Nicole M A Blijlevens
- Radboud Institute of Health Sciences, Department of Hematology, Radboud University Medical Center, PO Box 9101, 6500, HB, Nijmegen, the Netherlands
| |
Collapse
|
32
|
Hanindita MH, Irawan R, Ugrasena IDG, Hariastawa IGBA. Comparison of two lipid emulsions on interleukin-1β, interleukin-8 and fatty acid composition in infants post gastrointestinal surgery: a randomized trial. F1000Res 2020; 9:1168. [PMID: 33299555 PMCID: PMC7707114 DOI: 10.12688/f1000research.26269.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Nutritional support plays an essential role for recovery in infants who undergo gastrointestinal surgery. The current standard type of intravenous lipid emulsion (IVLE) used as parenteral nutrition is the mixture of medium-chain triglyceride (MCT) and long chain triglyceride (LCT) rich in ω-6. Studies showed that ω-6 is associated with higher level of proinflammatory cytokines, leading to increased mortality rate, morbidity rate, and postoperative recovery time. The latest generation of emulsion is a mixture of MCT, LCT, olive oil (OO), and fish oil (FO) which may optimize the ω6/ω3 ratio. This study aimed to compare the effect of MCT/LCT/OO/FO IVLE to standard IVLE on IL-1β, IL-8 and serum fatty acids in infants who had undergone gastrointestinal surgery. Methods: A single-blind, randomised controlled, pretest-posttest design study was done in twelve subjects that were classified into two groups. Group 1 received standard IVLE, group 2 received MCT/LCT/OO/FO IVLE. The type of standard and MCT/LCT/OO/FO IVLE used in this study were Lipofundin 20% and SMOFlipid 20%, respectively, both administered for three consecutive days in 1-4 gram/kilogram/day. IL-1β and IL-8 were examined using ELISA while fatty acids was analyzed using gas chromatography tandem mass spectrometry (GC-MS). Statistical analyses were performed using SPSS for Mac 23. Results: No statistical difference was found in age, gender, birth weight and diagnosis between both groups. Leukocyte was significantly lower in MCT/LCT/OO/FO group 3 days after surgery (p=0.025). CRP was lower in MCT/LCT/OO/FO group 3 days after surgery (p=0.01) and in changes within 3 days (p=0.016). There were no differences in IL-1β, IL-8 and ω-3 but ω-6 was higher in standard IVFE group on third day after surgery (p=0,048) Conclusion: MCT/LCT/OO/FO IVLE can significantly lower leukocyte, CRP and ω-6 levels and is comparable with standard IVLE on IL-1β, IL-8 and ω-3 levels in infants who had undergone gastrointestinal surgery.
Collapse
Affiliation(s)
- Meta Herdiana Hanindita
- Doctoral Program, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - Roedi Irawan
- Department of Child Health, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - I Dewa Gede Ugrasena
- Department of Child Health, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| | - I G B Adria Hariastawa
- Department of Pediatric Surgery, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, 60286, Indonesia
| |
Collapse
|
33
|
Shukla S, Tripathi AK, Verma SP, Yadav DK, Tripathi RK, Maurya S, Awasthi N. Association of Interleukin-1β-31C/T, -511T/C and -3954C/T Single Nucleotide Polymorphism and Their Blood Plasma Level in Acquired Aplastic Anemia. Indian J Hematol Blood Transfus 2020; 37:210-219. [PMID: 33867726 DOI: 10.1007/s12288-020-01281-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/15/2020] [Indexed: 11/30/2022] Open
Abstract
Aplastic anemia (AA) is an immune-mediated disorder in which hematopoietic stem and progenitor cells are targeted by a number of cellular and molecular pathways. This case control study aims to investigate the association of interleukin-1beta (IL-1β) gene polymorphisms, (IL-1β-31, IL-1β-511 and IL-1β-3954) and their plasma levels with acquired AA. Genotyping was done by Restricted Fragment Length Polymorphism (PCR-RFLP) method and IL-1β plasma levels were evaluated in peripheral blood using ELISA. Increased level of IL-1β was reported to be significant in cases as compared to controls. The susceptibility of developing AA was higher in the cases for IL-1β-3954 genotype. IL-1β-511 genotype showed significant association with the severity groups of AA. No significant association was noticed in responder versus non-responder group. Plasma level of IL-1β gene was found to be significantly higher in severe and very-severe group of AA versus control group. Our findings suggest that IL-1β gene and its genotypes might be involved in the pathophysiology of AA and play a central role in the etiopathogenesis of AA.
Collapse
Affiliation(s)
- Saurabh Shukla
- Department of Clinical Hematology, King George's Medical University, Chowk, Lucknow, 226003 India
| | - Anil Kumar Tripathi
- Department of Clinical Hematology, King George's Medical University, Chowk, Lucknow, 226003 India
| | - Shailendra Prasad Verma
- Department of Clinical Hematology, King George's Medical University, Chowk, Lucknow, 226003 India
| | - Deependra Kumar Yadav
- Department of Clinical Hematology, King George's Medical University, Chowk, Lucknow, 226003 India
| | - R K Tripathi
- Division of Toxicology, Central Drug Research Institute, Sitapur Road, Jankipuram Extension, Lucknow, 226031 India
| | - Shweta Maurya
- Department of Clinical Hematology, King George's Medical University, Chowk, Lucknow, 226003 India
| | - Nidhi Awasthi
- Department of Pediatrics, King George's Medical University, Chowk, Lucknow, 226003 India
| |
Collapse
|
34
|
Kim JB, Prunicki M, Haddad F, Dant C, Sampath V, Patel R, Smith E, Akdis C, Balmes J, Snyder MP, Wu JC, Nadeau KC. Cumulative Lifetime Burden of Cardiovascular Disease From Early Exposure to Air Pollution. J Am Heart Assoc 2020; 9:e014944. [PMID: 32174249 PMCID: PMC7335506 DOI: 10.1161/jaha.119.014944] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The disease burden associated with air pollution continues to grow. The World Health Organization (WHO) estimates ≈7 million people worldwide die yearly from exposure to polluted air, half of which-3.3 million-are attributable to cardiovascular disease (CVD), greater than from major modifiable CVD risks including smoking, hypertension, hyperlipidemia, and diabetes mellitus. This serious and growing health threat is attributed to increasing urbanization of the world's populations with consequent exposure to polluted air. Especially vulnerable are the elderly, patients with pre-existing CVD, and children. The cumulative lifetime burden in children is particularly of concern because their rapidly developing cardiopulmonary systems are more susceptible to damage and they spend more time outdoors and therefore inhale more pollutants. World Health Organization estimates that 93% of the world's children aged <15 years-1.8 billion children-breathe air that puts their health and development at risk. Here, we present growing scientific evidence, including from our own group, that chronic exposure to air pollution early in life is directly linked to development of major CVD risks, including obesity, hypertension, and metabolic disorders. In this review, we surveyed the literature for current knowledge of how pollution exposure early in life adversely impacts cardiovascular phenotypes, and lay the foundation for early intervention and other strategies that can help prevent this damage. We also discuss the need for better guidelines and additional research to validate exposure metrics and interventions that will ultimately help healthcare providers reduce the growing burden of CVD from pollution.
Collapse
Affiliation(s)
- Juyong Brian Kim
- Division of Cardiovascular MedicineDepartment of MedicineStanford UniversityStanfordCA
- Stanford Cardiovascular InstituteStanford UniversityStanfordCA
| | - Mary Prunicki
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| | - Francois Haddad
- Division of Cardiovascular MedicineDepartment of MedicineStanford UniversityStanfordCA
- Stanford Cardiovascular InstituteStanford UniversityStanfordCA
| | - Christopher Dant
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| | - Rushali Patel
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| | - Eric Smith
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| | - Cezmi Akdis
- Swiss Institute for Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - John Balmes
- Department of MedicineUniversity of California San Francisco and Division of Environmental Health SciencesSchool of Public HealthUniversity of California BerkeleyCA
| | - Michael P. Snyder
- Department of Genetics and Center for Genomics and Personalized MedicineStanford UniversityStanfordCA
| | - Joseph C. Wu
- Stanford Cardiovascular InstituteStanford UniversityStanfordCA
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma ResearchStanford UniversityStanfordCA
| |
Collapse
|
35
|
Garon EB, Chih-Hsin Yang J, Dubinett SM. The Role of Interleukin 1β in the Pathogenesis of Lung Cancer. JTO Clin Res Rep 2020; 1:100001. [PMID: 34589908 PMCID: PMC8474414 DOI: 10.1016/j.jtocrr.2020.100001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Chronic inflammation is associated with an increased risk of several diseases, including cancer. A complex tumor microenvironment created and maintained by a range of cell types promotes tumor growth, angiogenesis, and metastasis. Inflammasomes, multicomplex cytosolic proteins, generate much of this inflammation, including the activation of the cytokine interleukin (IL)-1β. Inflammation generated by IL-1β is present in several disease states, including atherosclerosis, diabetes, and arthritis. IL-1β is activated when a specific inflammasome, nucleotide-binding domain-like receptor protein 3, induces cleavage of pro-IL-1β into its active form. Nucleotide-binding domain-like receptor protein 3 is up-regulated in lung cancer; IL-1β binds to its receptor and activates signaling pathways, including the MAPK, cyclooxygenase, and nuclear factor-κB pathways, leading to macrophage activation, intratumoral accumulation of immunosuppressive myeloid cells, and tumor growth, invasiveness, metastasis, and angiogenesis. Evidence suggests a role for IL-1β and some of its downstream effectors (e.g., IL-6, IL-8, C-reactive protein, cyclooxygenase-2) as prognostic markers in many malignancies, including lung cancer. METHODS A phase III cardiovascular study of canakinumab, a human immunoglobulin Gk monoclonal antibody with high affinity and specificity for IL-1β, was conducted in patients who had a myocardial infarction. RESULTS A subanalysis of this study found that treatment with canakinumab substantially reduced incident lung cancer and lung cancer mortality in a dose-dependent manner. CONCLUSIONS A phase III trial is currently recruiting participants to evaluate canakinumab as adjuvant treatment versus placebo in patients with lung cancer. Other studies are investigating combinations of established antineoplastic agents and canakinumab in both early- and advanced-stage NSCLC.
Collapse
Affiliation(s)
- Edward B. Garon
- Division of Hematology and Oncology, David Geffen School of Medicine at UCLA (TRIO-US Network), Santa Monica, California
| | - James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Steven M. Dubinett
- Departments of Medicine, Pathology, and Laboratory Medicine and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
| |
Collapse
|
36
|
|
37
|
Wang Y, Sun X, Yuan S, Hou S, Guo T, Chu Y, Pang T, Luo HR, Yuan W, Wang X. Interleukin-1β inhibits normal hematopoietic expansion and promotes acute myeloid leukemia progression via the bone marrow niche. Cytotherapy 2020; 22:127-134. [PMID: 32024607 DOI: 10.1016/j.jcyt.2020.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Abstract
Enhanced interleukin-1β (IL-1β) signaling is a common event in patients with acute myeloid leukemia (AML). It was previously demonstrated that chronic IL-1β exposure severely impaired hematopoietic stem cell (HSC) self-renewal capability in mice and promoted leukemia cell growth in primary AML cells. However, the role of IL-1β in the murine bone marrow (BM) niche remains unclear. Here, we explored the role of IL-1β in the BM niche in Il-1r1-/- mice, chronic IL-1β exposure mice and mixed lineage leukemia-AF9 fusion gene (MLL-AF9)-induced AML mice models. We demonstrated that IL-1R1 deficiency did not affect the function of HSCs or niche cells under steady-state conditions or during transplantation. Chronic exposure to IL-1β decreased the expansion of Il-1r1-/- hematopoietic cells in Il-1r1+/+ recipient mice. These results indicated that IL-1β exposure impaired the ability of niche cells to support hematopoietic cells. Furthermore, we revealed that IL-1R1 deficiency in niche cells prolonged the survival of MLL-AF9-induced AML mice. The results of our study suggest that inhibition of the IL-1β/IL-1R1 signaling pathway in the niche might be a non-cell-autonomous therapy strategy for AML.
Collapse
Affiliation(s)
- Yuxia Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiaolu Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Department of Anesthesiology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Shengnan Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shuaibing Hou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tengxiao Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yajing Chu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tianxiang Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Hongbo R Luo
- Department of Pathology, Harvard Medical School, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Xiaomin Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| |
Collapse
|
38
|
Xu Z, Xie C, Xia L, Yuan Y, Zhu H, Huang X, Li C, Tao Y, Qu X, Zhang F, Zhang Z. Targeted exome sequencing identifies five novel loci at genome-wide significance for modulating antidepressant response in patients with major depressive disorder. Transl Psychiatry 2020; 10:30. [PMID: 32066657 PMCID: PMC7026085 DOI: 10.1038/s41398-020-0689-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/23/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
In order to determine the role of single nucleotide variants (SNVs) in modulating antidepressant response, we conducted a study, consisting of 929 major depressive disorder (MDD) patients, who were treated with antidepressant drugs (drug-only) or in combination with a repetitive transcranial magnetic stimulation (plus-rTMS), followed by targeted exome sequencing analysis. We found that the "plus-rTMS" patients presented a more effective response to the treatment when compared to the 'drug-only' group. Our data firstly demonstrated that the SNV burden had a significant impact on the antidepressant response presented in the "drug-only" group, but was limited in the "plus-rTMS" group. Further, after controlling for overall SNV burden, seven single nucleotide polymorphisms (SNPs) at five loci, IL1A, GNA15, PPP2CB, PLA2G4C, and GBA, were identified as affecting the antidepressant response at genome-wide significance (P < 5 × 10-08). Additional multiple variants achieved a level of correction for multiple testing, including GNA11, also shown as a strong signal for MDD risk. Our study showed some promising evidence on genetic variants that could be used as individualized therapeutic guides for MDD patients.
Collapse
Affiliation(s)
- Zhi Xu
- grid.263826.b0000 0004 1761 0489The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009 Nanjing, Jiangsu China
| | - Chunming Xie
- grid.263826.b0000 0004 1761 0489The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009 Nanjing, Jiangsu China
| | - Lu Xia
- Global Clinical and Translational Research Institute, Bethesda, MD 20814 USA
| | - Yonggui Yuan
- grid.263826.b0000 0004 1761 0489The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009 Nanjing, Jiangsu China
| | - Hong Zhu
- grid.263826.b0000 0004 1761 0489The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009 Nanjing, Jiangsu China
| | - Xiaofa Huang
- grid.263826.b0000 0004 1761 0489The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009 Nanjing, Jiangsu China
| | - Caihua Li
- Center for Genetics and Genomics Analysis, Genesky Biotechnologies, Inc, 201203 Shanghai, China
| | - Yu Tao
- Center for Genetics and Genomics Analysis, Genesky Biotechnologies, Inc, 201203 Shanghai, China
| | - Xiaoxiao Qu
- Genesky Diagnostics, Inc., BioBay, SIP, 215123 Jiangsu, China
| | - Fengyu Zhang
- Global Clinical and Translational Research Institute, Bethesda, MD, 20814, USA.
| | - Zhijun Zhang
- The Department of Neurology and Psychiatry of Affiliated ZhongDa Hospital, and Medical School of Southeast University, 210009, Nanjing, Jiangsu, China. .,Global Clinical and Translational Research Institute, Bethesda, MD, 20814, USA. .,The Institute of Neuropsychiatry, the Key Laboratory of Development Genes and Human Diseases, the Ministry of Education and Institute of Life Sciences of Southeast University, 210096, Nanjing, Jiangsu, China.
| |
Collapse
|
39
|
He C, Zhang Y, Luo H, Luo B, He Y, Jiang N, Liang Y, Zeng J, Luo Y, Xian Y, Liu J, Zheng X. Identification of the key differentially expressed genes and pathways involved in neutrophilia. Innate Immun 2019; 26:270-284. [PMID: 31726910 PMCID: PMC7251796 DOI: 10.1177/1753425919887411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the most important determinants in the acute inflammatory response. Pathologically increased numbers of PMNs in the circulation or specific tissues (or both) lead to neutrophilia. However, the genes expressed and pathways involved in neutrophilia have yet to be elucidated. By analysis of three public microarray datasets related to neutrophilia (GSE64457, GSE54644, and GSE94923) and evaluation by gene ontology, pathway enrichment, protein-protein interaction networks, and hub genes analysis using multiple methods (DAVID, PATHER, Reactome, STRING, Reactome FI Plugin, and CytoHubba in Cytoscape), we identified the commonly up-regulated and down-regulated different expressed genes. We also discovered that multiple signaling pathways (IL-mediated, LPS-mediated, TNF-α, TLR cascades, MAPK, and PI3K-Akt) were involved in PMN regulation. Our findings suggest that the commonly expressed genes involved in regulation of multiple pathways were the underlying molecular mechanisms in the development of inflammatory, autoimmune, and hematologic diseases that share the common phenotypic characteristics of increased numbers of PMNs. Taken together, these data suggest that these genes are involved in the regulation of neutrophilia and that the corresponding gene products could serve as potential biomarkers and/or therapeutic targets for neutrophilia.
Collapse
Affiliation(s)
- Chengcheng He
- People's Hospital of Zhongjiang, Deyang, Sichuan, P. R. China.,College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Yingchun Zhang
- People's Hospital of Zhongjiang, Deyang, Sichuan, P. R. China.,College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Hongwei Luo
- People's Hospital of Mianzhu, Deyang, Sichuan, P. R. China
| | - Bo Luo
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Yancheng He
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Nan Jiang
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Yu Liang
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Jingyuan Zeng
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Yujiao Luo
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Yujun Xian
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Jiajia Liu
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| | - Xiaoli Zheng
- College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, P. R. China
| |
Collapse
|
40
|
Aydın M. A sensitive and selective approach for detection of IL 1α cancer biomarker using disposable ITO electrode modified with epoxy-substituted polythiophene polymer. Biosens Bioelectron 2019; 144:111675. [DOI: 10.1016/j.bios.2019.111675] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/26/2019] [Accepted: 09/03/2019] [Indexed: 01/05/2023]
|
41
|
Eyre R, Alférez DG, Santiago-Gómez A, Spence K, McConnell JC, Hart C, Simões BM, Lefley D, Tulotta C, Storer J, Gurney A, Clarke N, Brown M, Howell SJ, Sims AH, Farnie G, Ottewell PD, Clarke RB. Microenvironmental IL1β promotes breast cancer metastatic colonisation in the bone via activation of Wnt signalling. Nat Commun 2019; 10:5016. [PMID: 31676788 PMCID: PMC6825219 DOI: 10.1038/s41467-019-12807-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023] Open
Abstract
Dissemination of tumour cells to the bone marrow is an early event in breast cancer, however cells may lie dormant for many years before bone metastases develop. Treatment for bone metastases is not curative, therefore new adjuvant therapies which prevent the colonisation of disseminated cells into metastatic lesions are required. There is evidence that cancer stem cells (CSCs) within breast tumours are capable of metastasis, but the mechanism by which these colonise bone is unknown. Here, we establish that bone marrow-derived IL1β stimulates breast cancer cell colonisation in the bone by inducing intracellular NFkB and CREB signalling in breast cancer cells, leading to autocrine Wnt signalling and CSC colony formation. Importantly, we show that inhibition of this pathway prevents both CSC colony formation in the bone environment, and bone metastasis. These findings establish that targeting IL1β-NFKB/CREB-Wnt signalling should be considered for adjuvant therapy to prevent breast cancer bone metastasis.
Collapse
Affiliation(s)
- Rachel Eyre
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Denis G Alférez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Angélica Santiago-Gómez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Kath Spence
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - James C McConnell
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Claire Hart
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Bruno M Simões
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Diane Lefley
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Claudia Tulotta
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - Joanna Storer
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Austin Gurney
- OncoMed Pharmaceuticals, Redwood City, CA, 94063, USA
| | - Noel Clarke
- Department of Urology, Salford Royal Hospital NHS Foundation Trust, Stott Lane, Salford, M6 8HD, UK
| | - Mick Brown
- Genito Urinary Cancer Research Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer Group, Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Gillian Farnie
- Structural Genomics Consortium, NDORMS, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK
| | - Penelope D Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK.
| |
Collapse
|
42
|
Liu WH, Shi LS, Chung MC, Chang TC, Lee SY. Antcamphin M Inhibits TLR4-Mediated Inflammatory Responses by Upregulating the Nrf2/HO-1 Pathway and Suppressing the NLRP3 Inflammasome Pathway in Macrophages. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1611-1626. [PMID: 31645125 DOI: 10.1142/s0192415x19500824] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The medicinal mushroom Antrodia cinnamomea has been demonstrated to have anti-inflammatory properties. However, the bioactive compounds in A. cinnamomea need further investigation. The present study aimed to understand the mechanism of action of antcamphin M, an ergostanoid isolated from A. cinnamomea mycelium and to clarify its underlying mechanisms of action. RAW264.7 cells were pretreated with the indicated concentrations of antcamphin M, prior to stimulation with lipopolysaccharide (LPS). Cell viability, production of nitric oxide (NO), prostaglandin E2 (PGE2), cytokines, and chemokines, as well as the inflammation-related signaling pathways were investigated. The study revealed that antcamphin M significantly decreased the LPS-induced production of NO, PGE2, pro-inflammatory cytokines, and keratinocyte chemoattractant CXCL1 (KC), along with the levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) proteins without significant cytotoxicity, indicating it had a better anti-inflammatory activity than that of gisenoside Rb1 and Rg1. Additionally, antcamphin M significantly inhibited the activation of MAPKs (p38, ERK, and JNK), NFκB, and components of the NLRP3 inflammasome (NLRP3, ASC, and caspase-1) signaling pathways and also increased the levels of nuclear factor erythroid-2-related factor (Nrf2) and heme oxygenase-1 (HO-1). These findings suggest that antcamphin M possesses potent anti-inflammatory activities and could be a potential candidate for the development of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Wei-Hsiu Liu
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Li-Shian Shi
- Department of Biotechnology, National Formosa University, Yunlin, Taiwan
| | - Min-Chieh Chung
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Tsu-Chung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Yu Lee
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
43
|
Benner B, Scarberry L, Stiff A, Duggan MC, Good L, Lapurga G, Butchar JP, Tridandapani S, Carson WE. Evidence for interaction of the NLRP3 inflammasome and Bruton's tyrosine kinase in tumor-associated macrophages: implications for myeloid cell production of interleukin-1beta. Oncoimmunology 2019; 8:1659704. [PMID: 31646085 PMCID: PMC6791459 DOI: 10.1080/2162402x.2019.1659704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/05/2023] Open
Abstract
An inflammatory microenvironment has been shown to play an important role in the growth and metastasis of tumors. The NLRP3 inflammasome is a multi-protein complex of the innate immune system that is responsible for the production of the potent inflammatory cytokine IL-1β. Tumor- associated macrophages (TAM) are an expanded population of immune cells found in the tumor microenvironment that can promote the initiation and metastasis of tumor cells. Their presence has been correlated with disease burden, highlighting the therapeutic potential of targeting this population. However, to date clinically relevant pharmacologic strategies to target TAM remain elusive. Here, we show that in vitro generated TAM harbor NLRP3 inflammasome components and produce IL-1β. Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK), is in clinical use for the treatment of B- cell malignancies. We report that BTK is expressed by human in vitro generated TAM and murine macrophages and that it physically associates with the NLRP3 inflammasome. Furthermore, ibrutinib is able to inhibit BTK phosphorylation in TAM generated in vitro. Treatment of TAM with ibrutinib significantly impaired the ability of these cells to produce IL-1β. The present study provides evidence that BTK physically associates with the NLRP3 inflammasome and that inhibition of BTK with ibrutinib can impair the production of IL-1β by in vitro generated TAM. Thus, ibrutinib could potentially be of clinical use in abrogating inflammation-associated cancer progression and the immune-suppressive effects of myeloid cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Luke Scarberry
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Andrew Stiff
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Megan C. Duggan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Logan Good
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Gabriella Lapurga
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | | | - William E. Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Division of Surgical Oncology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
44
|
Zhu X, Wang Y, Jiang Q, Jiang H, Lu J, Wang Y, Kong Y, Chang Y, Xu L, Peng J, Hou M, Huang X, Zhang X. All- trans retinoic acid protects mesenchymal stem cells from immune thrombocytopenia by regulating the complement-interleukin-1β loop. Haematologica 2019; 104:1661-1675. [PMID: 30679324 PMCID: PMC6669169 DOI: 10.3324/haematol.2018.204446] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
Enhanced peripheral complement activation has long been considered as one of the major pathogenic elements of immune thrombocytopenia. A dysfunctional bone marrow microenvironment, especially with regards to mesenchymal stem cells, has been observed in patients with immune thrombocytopenia. However, the potential role of the complement system in the dysfunctional bone marrow microenvironment remains poorly understood. In this study, bone marrow samples from patients with immune thrombocytopenia were divided into two groups based on whether or not complement components were deposited on the surfaces of their mesenchymal stem cells. The mesenchymal cells from the group with complement deposition were less numerous, dysfunctional, had a reduced capacity to proliferate, and showed increased apoptosis as well as abnormal secretion of interleukin-1β and C-X-C motif chemokine ligand 12. In vitro treatment with all-trans retinoic acid increased the number and improved the function of the complement-positive bone marrow mesenchymal stem cells by upregulating DNA hypermethylation of the interleukin-1β promoter. In vivo studies showed that all-trans retinoic acid could rescue the impaired mesenchymal stem cells to support the thrombopoietic niche in both patients with immune thrombocytopenia and a murine model of this disease. Taken together, these results indicate that impairment of mesenchymal stem cells, mediated by the complement-interleukin-1β loop, plays a role in the pathogenesis of immune thrombocytopenia. All-trans retinoic acid represents a promising therapeutic approach in patients with immune thrombocytopenia through its effect of repairing mesenchymal stem cell impairment.
Collapse
Affiliation(s)
- Xiaolu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yanan Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Hao Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Jin Lu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yazhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yuan Kong
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, Beijing
| |
Collapse
|
45
|
Anti-Tumor Effects of Vitamin B2, B6 and B9 in Promonocytic Lymphoma Cells. Int J Mol Sci 2019; 20:ijms20153763. [PMID: 31374832 PMCID: PMC6696026 DOI: 10.3390/ijms20153763] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic inflammation can lead to tumour initiation and progression. Vitamin B complex has the ability to regulate the immune response and, therefore, inflammation but many of the mechanistic and molecular processes involved in this regulation are still not fully understood. This study sought to determine some of these processes by studying the effects of vitamin B2 (riboflavin) B6 (pyridoxine) and B9 (folic acid) on un-differentiated pro-monocytic lymphoma cells in regard to their ability to alter the proliferation, migration, apoptosis, cytokines and expression levels of programmed death ligand 1. We show that vitamin B2, B6 and B9, on pro-monocytic lymphoma cells exerted an anti-tumorigenic effect. This data could form the basis for future studies in using vitamin B supplementation to reduce cancer cell growth in vivo.
Collapse
|
46
|
Van Gorp H, Lamkanfi M. The emerging roles of inflammasome-dependent cytokines in cancer development. EMBO Rep 2019; 20:embr.201847575. [PMID: 31101676 DOI: 10.15252/embr.201847575] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/02/2019] [Accepted: 04/30/2019] [Indexed: 12/24/2022] Open
Abstract
In addition to the genomic alterations that occur in malignant cells, the immune system is increasingly appreciated as a critical axis that regulates the rise of neoplasms and the development of primary tumours and metastases. The interaction between inflammatory cell infiltrates and stromal cells in the tumour microenvironment is complex, with inflammation playing both pro- and anti-tumorigenic roles. Inflammasomes are intracellular multi-protein complexes that act as key signalling hubs of the innate immune system. They respond to cellular stress and trauma by promoting activation of caspase-1, a protease that induces a pro-inflammatory cell death mode termed pyroptosis along with the maturation and secretion of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Here, we will briefly introduce inflammasome biology with a focus on the dual roles of inflammasome-produced cytokines in cancer development. Despite emerging insight that inflammasomes may promote and suppress cancer development according to the tumour stage and the tumour microenvironment, much remains to be uncovered. Further exploration of inflammasome biology in tumorigenesis should enable the development of novel immunotherapies for cancer patients.
Collapse
Affiliation(s)
- Hanne Van Gorp
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Center for Inflammation Research, VIB, Ghent, Belgium
| | - Mohamed Lamkanfi
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium .,Janssen Immunosciences, World Without Disease Accelerator, Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium
| |
Collapse
|
47
|
Ying Z, Shiue L, Park K, Kollet J, Bijani P, Goswami M, Duvic M, Ni X. Blood transcriptional profiling reveals IL-1 and integrin signaling pathways associated with clinical response to extracorporeal photopheresis in patients with leukemic cutaneous T-cell lymphoma. Oncotarget 2019; 10:3183-3197. [PMID: 31139332 PMCID: PMC6516711 DOI: 10.18632/oncotarget.26900] [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: 03/01/2019] [Accepted: 04/14/2019] [Indexed: 01/07/2023] Open
Abstract
Extracorporeal photopheresis (ECP) is a frontline therapy for patients with leukemic cutaneous T-cell lymphoma (L-CTCL), but its mechanisms of action are not fully understood. This study was to explore the molecular mechanisms underlying clinical response versus non-response in patients with L-CTCL. We performed blood transcriptional profiling of ten L-CTCL patients at Day 2 and 1 month post- ECP compared to pre-ECP baseline using Agilent Whole Human Genome Microarray technology. Differentially expressed genes (DEGs) between five clinically-responsive patients and five clinically-resistant patients were cross-compared. Higher numbers of genes were modulated in responders than non-responders after ECP at both Day 2 and 1 month, with two thirds of DEGs down-regulated. The down-regulated DEGs at 1 month post-ECP were related to inflammatory, immune and/or stress responses, platelet functions, and chromatin remodeling. Upregulated DEGs were mainly related to functions of the nucleolus. Pathway analysis revealed that integrin and IL-1 signaling pathways were the top pathways affected in responders, which were minimally affected in non-responders. The top upstream transcription regulators affected were IL1B, EGR1, FAS, and TGFB1. Our results suggest that the modulation of cell adhesion and suppression of IL-1β induced inflammation may underlie the efficacy of ECP in L-CTCL.
Collapse
Affiliation(s)
- Zuolin Ying
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lisa Shiue
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katherine Park
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jutta Kollet
- Bioinformatics, Miltenyi Biotec GmbH, Beigisch Gladbach, 51429, Germany
| | - Pedram Bijani
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Meghali Goswami
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Madeleine Duvic
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiao Ni
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
48
|
Wang Y, Zuo X. Cytokines frequently implicated in myeloproliferative neoplasms. Cytokine X 2019; 1:100005. [PMID: 33604548 PMCID: PMC7885877 DOI: 10.1016/j.cytox.2019.100005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
MPN is a chronic inflammation-driven tumor model. Many cytokines are involved in pathogenesis and progression of MPN. IL-1β, TNF-α, IL-6, IL-8, VEGF, PDGF, TGF-β and IFNs are critical in MPN. Cytokine directed therapy could be an alternative treatment for MPN in future.
Classical myeloproliferative neoplasms (MPN) include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). MPN has been defined as a chronic inflammation-driven tumor model. It is clear that there is a close link between chronic inflammation and MPN pathogenesis. Several studies have demonstrated cytokine profiles in MPN patients. Other studies have used cell lines or animal models aiming to clarify the underlying mechanism of cytokines in the pathogenesis of MPN. However, important questions remain: (1) among all these cytokines, which are more predictive? and (2) which are more critical? In this review, we summarize cytokines that have been investigated in MPN and highlight several cytokines that may be more significant in MPN. We suggest that cytokines are more critical in PMF than PV or ET. These cytokines include IL-1β, TNF-α, IL-6, IL-8, VEGF, PDGF, IFNs and TGF-β, all of which should be more closely investigated in MPN. Based on our extensive literature search, several key factors have emerged in our understanding of MPN: first, TNF-α could correlate with MPN progression including PMF, PV and ET. IL-1β plays a role in PMF progression, while it showed no relation with PV or ET. Second, IL-8 could be a prognostic factor for PMF, and IL-6 could be important for MPN progression. Third, VEGF and PDGF play an indirect role in MPN development and their inhibitors could be effective. Fourth, different subtypes of IFNs could have different effects in MPN. Finally, TGF-β is closely linked to MF, although the data are inconsistent. Agents that have targeted these cytokines described above are already in clinical trials, and some of them have even been used to treat MPN patients. Taken together, it will be critical to continue to investigate the precise role of these cytokines in the pathogenesis and progression of MPN.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, Hubei 430071, PR China
| | - Xuelan Zuo
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, Hubei 430071, PR China
| |
Collapse
|
49
|
Nalle SC, Zuo L, Ong MLDM, Singh G, Worthylake AM, Choi W, Manresa MC, Southworth AP, Edelblum KL, Baker GJ, Joseph NE, Savage PA, Turner JR. Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability. J Clin Invest 2019; 129:902-914. [PMID: 30667372 PMCID: PMC6355225 DOI: 10.1172/jci98554] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a complication of hematopoietic stem cell transplantation (HSCT) that affects multiple organs. GVHD-associated intestinal damage can be separated into two distinct phases, initiation and propagation, which correspond to conditioning-induced damage and effector T cell activation and infiltration, respectively. Substantial evidence indicates that intestinal damage induced by pretransplant conditioning is a key driver of GVHD initiation. Here, we aimed to determine the impact of dysregulated intestinal permeability on the subsequent GVHD propagation phase. The initiation phase of GVHD was unchanged in mice lacking long MLCK (MLCK210), an established regulator of epithelial tight junction permeability. However, MLCK210-deficient mice were protected from sustained barrier loss and exhibited limited GVHD propagation, as indicated by reduced histopathology, fewer CD8+ effector T cells in the gut, and improved overall survival. Consistent with these findings, intestinal epithelial MLCK210 expression and enzymatic activity were similarly increased in human and mouse GVHD biopsies. Intestinal epithelial barrier loss mediated by MLCK210 is therefore a key driver of the GVHD propagation. These data suggest that inhibition of MLCK210-dependent barrier regulation may be an effective approach to limiting GVHD progression.
Collapse
Affiliation(s)
- Sam C. Nalle
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Li Zuo
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Anhui Medical University, Hefei, Anhui, China
| | - Ma. Lora Drizella M. Ong
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gurminder Singh
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alicia M. Worthylake
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wangsun Choi
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mario Cabrero Manresa
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna P. Southworth
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Karen L. Edelblum
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Cancer Center, Newark, New Jersey, USA
| | - Gregory J. Baker
- Laboratory of Systems Pharmacology, Harvard Medical School, Harvard Program in Therapeutic Science, Boston, Massachusetts, USA
| | - Nora E. Joseph
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Peter A. Savage
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Pellegrini C, Fornai M, Colucci R, Benvenuti L, D’Antongiovanni V, Natale G, Fulceri F, Giorgis M, Marini E, Gastaldi S, Bertinaria M, Blandizzi C, Antonioli L. A Comparative Study on the Efficacy of NLRP3 Inflammasome Signaling Inhibitors in a Pre-clinical Model of Bowel Inflammation. Front Pharmacol 2018; 9:1405. [PMID: 30559669 PMCID: PMC6287041 DOI: 10.3389/fphar.2018.01405] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022] Open
Abstract
Nucleotide-binding oligomerization domain leucine rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome is pivotal in maintaining intestinal homeostasis and sustaining enteric immune responses in the setting of inflammatory bowel diseases. Drugs acting as NLRP3 blockers could represent innovative strategies for treatment of bowel inflammation. This study was performed in rats with dinitrobenzenesulfonic acid (DNBS)-induced colitis, to investigate how the direct blockade of NLRP3 inflammasome with an irreversible inhibitor (INF39) compares with Ac-YVAD-cmk (YVAD, caspase-1 inhibitor) and anakinra (IL-1β receptor antagonist), acting downstream on NLRP3 signaling. Animals with DNBS-colitis received YVAD (3 mg/kg) or anakinra (100 mg/Kg) intraperitoneally, and INF39 (25 mg/kg) or dexamethasone (DEX, 1 mg/kg) orally for 6 days, starting on the same day of colitis induction. Under colitis, there was a body weight decrease, which was attenuated by YVAD, anakinra or INF39, but not DEX. All test drugs counteracted the increase in spleen weight. The colonic shortening and morphological colonic alterations associated with colitis were counteracted by INF39, anakinra and DEX, while YVAD was without effects. Tissue increments of myeloperoxidase, tumor necrosis factor and interleukin-1β were more effectively counteracted by INF39 and DEX, than YVAD and anakinra. These findings indicate that: (1) direct inhibition of NLRP3 inflammasome with INF39 is more effective than caspase-1 inhibition or IL-1β receptor blockade in reducing systemic and bowel inflammatory alterations; (2) direct NLRP3 inhibition can be a suitable strategy for treatment of bowel inflammation.
Collapse
Affiliation(s)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Federica Fulceri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Marta Giorgis
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Elisabetta Marini
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Simone Gastaldi
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Massimo Bertinaria
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|