1
|
Wu W, Chen Z, Wen H, Zhang H. Unveiling potential drug targets for lung squamous cell carcinoma through the integration of druggable genome and genome-wide association data. Front Genet 2024; 15:1431684. [PMID: 39175755 PMCID: PMC11338847 DOI: 10.3389/fgene.2024.1431684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Background: Lung squamous cell carcinoma (LSCC) is a major subtype of lung cancer with poor prognosis and low survival rate. Compared with lung adenocarcinoma, yet no FDA-approved targeted-therapy has been found for lung squamous cell carcinoma. Methods: To identify potential drug targets for LSCC, Summary-data-based Mendelian randomization (SMR) analysis was used to examine the potential association between 4,543 druggable genes and LSCC, followed by colocalization analysis and HEIDI tests to confirm the robustness of the result. Phenome-wide association study (PheWAS) explored potential side effects of candidate drug targets. Enrichment analysis and protein-protein interaction networks revealed the function and significance of therapeutic targets. Single-cell expression analysis was used to examine cell types with enrichment expression of druggable genes in LSCC tissue. Drug prediction included screening potential drug candidates and evaluating their interactions with targets through molecular docking. Results: This research has identified ten significant drug targets for LSCC through a comprehensive SMR analysis. These targets included (COPA, PKD2L1, CCR1, C2, CYP21A2, and NCSTN as risk factors, and CCNA2, C4A, APOM, and LPAR2 as protective factors). PheWAS demonstrated that C2, CCNA2, LPAR2, and NCSTN exhibited associations with other phenotypes at the genetic level. Then, we found four potentially effective drugs with the Dsigdb database. Subsequently, molecular docking indicated that favorable binding interactions between drug candidates and potential target molecules. In the druggability evaluation, five out of ten drug target genes have been used in drug development (APOM, C4A, CCNA2, COPA, and PKD2L1). Six out of ten druggable genes showed significant expression in LSCC tissues (COPA, PKD2L1, CCR1, C2, NCSTN, LPAR2). Besides, Single-cell expression analysis revealed that C2 and CCNA2 were primarily enriched in macrophages, while COPA and NCSTN were enriched in both macrophages and epithelial cells. Conclusion: Our research revealed ten potential druggable genes for LSCC treatment, which might help to advance the precise and efficient therapeutic approaches of LSCC.
Collapse
Affiliation(s)
- Wenhua Wu
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhengrui Chen
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiteng Wen
- The Second Clinical Medical College, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haiyun Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hosptial, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Di Donato R, Bonecchi R, Albano F. Canonical and atypical chemokine receptors in the neutrophil life cycle. Cytokine 2023; 169:156297. [PMID: 37453326 DOI: 10.1016/j.cyto.2023.156297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Chemokines are mainly studied for their local function in the control of leukocyte extravasation in homeostatic and inflammatory conditions. However, they have additional roles at the systemic level including the regulation of the hematopoietic process and leukocyte differentiation. Due to the redundancy and pleiotropicity of the chemokine system, chemokines have often multiple and complex roles in neutrophil differentiation ranging from retention and control of proliferation of progenitors to the mobilization of mature cells from the bone marrow (BM) to the bloodstream and their further differentiation in tissues. Atypical chemokine receptors (ACKRs) are regulators of the chemokine system by controlling chemokine bioavailability and chemokine receptor function. Even though ACKRs bind a wide range of chemokines, they appear to have a selective role in the process of neutrophil production and differentiation. The aim of this review is to give an overview of the current evidence regarding the role of chemokines and chemokine receptors in the life of neutrophils with a focus on the regulation exerted by ACKRs.
Collapse
Affiliation(s)
- Rachele Di Donato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| | - Raffaella Bonecchi
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy.
| | - Francesca Albano
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
| |
Collapse
|
3
|
Wojtowicz EE, Mistry JJ, Uzun V, Hellmich C, Scoones A, Chin DW, Kettyle LM, Grasso F, Lord AM, Wright DJ, Etherington GJ, Woll PS, Belderbos ME, Bowles KM, Nerlov C, Haerty W, Bystrykh LV, Jacobsen SEW, Rushworth SA, Macaulay IC. Panhematopoietic RNA barcoding enables kinetic measurements of nucleate and anucleate lineages and the activation of myeloid clones following acute platelet depletion. Genome Biol 2023; 24:152. [PMID: 37370129 PMCID: PMC10294477 DOI: 10.1186/s13059-023-02976-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Platelets and erythrocytes constitute over 95% of all hematopoietic stem cell output. However, the clonal dynamics of HSC contribution to these lineages remains largely unexplored. RESULTS We use lentiviral genetic labeling of mouse hematopoietic stem cells to quantify output from all lineages, nucleate, and anucleate, simultaneously linking these with stem and progenitor cell transcriptomic phenotypes using single-cell RNA-sequencing. We observe dynamic shifts of clonal behaviors through time in same-animal peripheral blood and demonstrate that acute platelet depletion shifts the output of multipotent hematopoietic stem cells to the exclusive production of platelets. Additionally, we observe the emergence of new myeloid-biased clones, which support short- and long-term production of blood cells. CONCLUSIONS Our approach enables kinetic studies of multi-lineage output in the peripheral blood and transcriptional heterogeneity of individual hematopoietic stem cells. Our results give a unique insight into hematopoietic stem cell reactivation upon platelet depletion and of clonal dynamics in both steady state and under stress.
Collapse
Affiliation(s)
- Edyta E Wojtowicz
- Earlham Institute, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Jayna J Mistry
- Earlham Institute, Norwich Research Park, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Vladimir Uzun
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Charlotte Hellmich
- Norwich Medical School, University of East Anglia, Norwich, UK
- Norfolk and Norwich University Hospital, Norwich, UK
| | - Anita Scoones
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Desmond W Chin
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Laura M Kettyle
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Francesca Grasso
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Allegra M Lord
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Petter S Woll
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Kristian M Bowles
- Norwich Medical School, University of East Anglia, Norwich, UK
- Norfolk and Norwich University Hospital, Norwich, UK
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Wilfried Haerty
- Earlham Institute, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Leonid V Bystrykh
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center of Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Sten Eirik W Jacobsen
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
- Department of Medicine, Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| | | | - Iain C Macaulay
- Earlham Institute, Norwich Research Park, Norwich, UK.
- Norwich Medical School, University of East Anglia, Norwich, UK.
| |
Collapse
|
4
|
Georgiou AC, Twisk JWR, Crielaard W, Ouwerling P, Schoneveld AH, van der Waal SV. The influence of apical periodontitis on circulatory inflammatory mediators in peripheral blood: A prospective case-control study. Int Endod J 2023; 56:130-145. [PMID: 36284462 PMCID: PMC10098678 DOI: 10.1111/iej.13854] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 01/17/2023]
Abstract
AIM To explore the influence of apical periodontitis (AP) on inflammatory markers in blood of otherwise healthy individuals and to depict the inflammatory profile of the healing after dental extraction. METHODOLOGY This is a prospective case-control intervention study, during which, individuals with a diagnosis of AP of one affected tooth were included, along with a control group matched for age and gender. A broad panel of blood inflammatory mediators was examined longitudinally in all subjects during six visits. In the case of the AP subjects, the tooth with AP was extracted at the third visit. Results were analysed by linear regression analyses and linear mixed-model analyses. RESULTS A total of 53 subjects were included in the study, 27 with AP and 26 without. Fifteen females and 12 males were included in the AP group, and 14 females and 12 males in the control group. At baseline, granulocyte colony-stimulating factor (p < .001), interleukin (IL)-1β (p = .03) and IL-4 (p = .01) were significantly lower in AP subjects than in controls. Comparison of the differences between baseline and the last visit, i.e. 3 months after the tooth extraction, showed a significant reduction in IL-10 (p = .03) and IL-12p70 (p = .01). CONCLUSIONS The immunologic profile of chronic AP in one tooth and its healing profile reveals a systemic low-grade inflammation through compensatory immunosuppression. A larger lesion or multiple lesions could disrupt the balance that the system is trying to maintain, resulting in loss of homeostasis.
Collapse
Affiliation(s)
- Athina Christina Georgiou
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Data Science, Amsterdam UMC, Location VU University Medical Center, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Wim Crielaard
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - Peter Ouwerling
- Tandheelkundig Centrum Molenvliet, Alphen aan den Rijn, The Netherlands
| | - Arjan H Schoneveld
- Division LAB, Department Central Diagnostic Laboratory, UMC Utrecht, Utrecht, The Netherlands
| | - Suzette Veronica van der Waal
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands.,Department of Endodontics, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Paudel S, Ghimire L, Jin L, Jeansonne D, Jeyaseelan S. Regulation of emergency granulopoiesis during infection. Front Immunol 2022; 13:961601. [PMID: 36148240 PMCID: PMC9485265 DOI: 10.3389/fimmu.2022.961601] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
During acute infectious and inflammatory conditions, a large number of neutrophils are in high demand as they are consumed in peripheral organs. The hematopoietic system rapidly responds to the demand by turning from steady state to emergency granulopoiesis to expedite neutrophil generation in the bone marrow (BM). How the hematopoietic system integrates pathogenic and inflammatory stress signals into the molecular cues of emergency granulopoiesis has been the subject of investigations. Recent studies in the field have highlighted emerging concepts, including the direct sensing of pathogens by BM resident or sentinel hematopoietic stem and progenitor cells (HSPCs), the crosstalk of HSPCs, endothelial cells, and stromal cells to convert signals to granulopoiesis, and the identification of novel inflammatory molecules, such as C/EBP-β, ROS, IL-27, IFN-γ, CXCL1 with direct effects on HSPCs. In this review, we will provide a detailed account of emerging concepts while reassessing well-established cellular and molecular players of emergency granulopoiesis. While providing our views on the discrepant results and theories, we will postulate an updated model of granulopoiesis in the context of health and disease.
Collapse
Affiliation(s)
- Sagar Paudel
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Laxman Ghimire
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Liliang Jin
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Duane Jeansonne
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Samithamby Jeyaseelan
- Center for Lung Biology and Disease, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Pathobiological Sciences, Louisiana State University (LSU) School of Veterinary Medicine, Baton Rouge, LA, United States.,Section of Pulmonary and Critical Care, Department of Medicine, LSU Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
6
|
Broxmeyer HE, Cooper SH, Ropa J. CXCL15/Lungkine has suppressive activity on proliferation and expansion of multi-potential, erythroid, granulocyte and macrophage progenitors in S-phase specific manner. Blood Cells Mol Dis 2021; 91:102594. [PMID: 34520986 PMCID: PMC9231597 DOI: 10.1016/j.bcmd.2021.102594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022]
Abstract
Cytokines/chemokines regulate hematopoiesis, most having multiple cell actions. Numerous but not all chemokine family members act as negative regulators of hematopoietic progenitor cell (HPC) proliferation, but very little is known about such effects of the chemokine, CXCL15/Lungkine. We found that CXCL15/Lungkine-/- mice have greatly increased cycling of multi cytokine-stimulated bone marrow and spleen hematopoietic progenitor cells (HPCs: CFU-GM, BFU-E, and CFU-GEMM) and CXCL15 is expressed in many bone marrow progenitor and other cell types. This suggests that CXCL15/Lungkine acts as a negative regulator of the cell cycling of these HPCs in vivo. Recombinant murine CXCL15/Lungkine, decreased numbers of functional HPCs during cytokine-enhanced ex-vivo culture of lineage negative mouse bone marrow cells. Moreover, CXCL15/Lungkine, through S-Phase specific actions, was able to suppress in vitro colony formation of normal wildtype mouse bone marrow CFU-GM, CFU-G, CFU-M, BFU-E, and CFU-GEMM. This clearly identifies the negative regulatory activity of CXCL15/Lungkine on proliferation of multiple types of mouse HPCs.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA.
| | - Scott H Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA
| | - James Ropa
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA
| |
Collapse
|
7
|
Ntanasis-Stathopoulos I, Fotiou D, Terpos E. CCL3 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1231:13-21. [PMID: 32060842 DOI: 10.1007/978-3-030-36667-4_2] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Within the tumor microenvironment, chemokines play a key role in immune cell trafficking regulation and immune landscape formulation. CCL3 or macrophage inflammatory protein-1α (MIP-1α), an important chemokine implicated in both immune surveillance and tolerance, has emerged as a prognostic biomarker in both solid and hematological malignancies. CCL3 exerts both antitumor and pro-tumor behavior which is context dependent highlighting the complexity of the underlying interrelated signaling cascades. Current CCL3-directed therapeutic approaches are investigational and further optimization is required to increase efficacy and minimize adverse events.
Collapse
Affiliation(s)
- Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Despoina Fotiou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
8
|
Bonavita O, Mollica Poeta V, Massara M, Mantovani A, Bonecchi R. Regulation of hematopoiesis by the chemokine system. Cytokine 2019; 109:76-80. [PMID: 29429849 DOI: 10.1016/j.cyto.2018.01.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/19/2018] [Accepted: 01/24/2018] [Indexed: 12/16/2022]
Abstract
Although chemokines are best known for their role in directing cell migration, accumulating evidence indicate their involvement in many other processes. This review focus on the role of chemokines in hematopoiesis with an emphasis on myelopoiesis. Indeed, many chemokine family members are an important component of the cytokine network present in the bone marrow that controls proliferation, retention, and mobilization of hematopoietic progenitors.
Collapse
Affiliation(s)
- Ornella Bonavita
- Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Valeria Mollica Poeta
- Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano (MI), Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20090 Pieve Emanuele (MI), Italy
| | - Matteo Massara
- Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano (MI), Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano (MI), Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20090 Pieve Emanuele (MI), Italy; The William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano (MI), Italy; Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20090 Pieve Emanuele (MI), Italy.
| |
Collapse
|
9
|
Quickly attainable and highly engrafting hematopoietic stem cells. BLOOD SCIENCE 2019; 1:113-115. [PMID: 35402793 PMCID: PMC8975002 DOI: 10.1097/bs9.0000000000000003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 11/26/2022] Open
|
10
|
Sun B, Lei Y, Cao Z, Zhou Y, Sun Y, Wu Y, Wang S, Guo W, Liu C. TroCCL4, a CC chemokine of Trachinotus ovatus, is involved in the antimicrobial immune response. FISH & SHELLFISH IMMUNOLOGY 2019; 86:525-535. [PMID: 30521967 DOI: 10.1016/j.fsi.2018.11.080] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 06/09/2023]
Abstract
CC chemokines are a large subfamily of chemokines that play an important role in the innate immune system. To date, several CC chemokines have been identified in fish species; however, the activities and functions of these putative chemokines remain ambiguous in teleosts, especially in the golden pompano, Trachinotus ovatus. Here, we characterized CC chemokine ligand 4 from T. ovatus (TroCCL4) and studied its functions. TroCCL4 contains a 294 bp open reading frame that encodes a putative peptide comprising 97 amino acids. TroCCL4 shares a high amino acid sequence similarity of 31.11%-78.35% with other CC chemokines sequences in humans and teleosts and has four cysteine residues that are conserved among other CC chemokines. TroCCL4 is also related to the macrophage inflammatory protein (MIP) group of CC chemokines. TroCCL4 expression was most abundant in immune organs and significantly upregulated in a time-dependent manner following Edwardsiella tarda infection. Recombinant TroCCL4 (rTroCCL4) induced the migration of peripheral blood leukocytes and the cellular proliferation of head kidney lymphocytes. In addition, rTroCCL4 inhibited the growth of Escherichia coli and E. tarda, indicating an antimicrobial function. Furthermore, the results of in vivo analysis showed that TroCCL4 overexpression in T. ovatus significantly enhanced macrophage activation; upregulated the gene expression of interleukin 1-β (IL-1β), interleukin 15 (IL15), interferon-induced Mx protein (Mx), tumor necrosis factor α (TNFα), complement C3, and major histocompatibility complex (MHC) class Iα and class IIα; and protected against bacterial infection in fish tissues. In contrast, knockdown of TroCCL4 expression resulted in increased bacterial dissemination and colonization in fish tissues. Taken together, our results provide evidence indicating that TroCCL4 has the ability to stimulate leukocytes and macrophages and enhance host immunity to defend against bacterial infection.
Collapse
Affiliation(s)
- Baiming Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China
| | - Yang Lei
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China
| | - Zhenjie Cao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, PR China
| | - Yongcan Zhou
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China
| | - Yun Sun
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China.
| | - Ying Wu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China
| | - Shifeng Wang
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China; Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China
| | - Weiliang Guo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, PR China
| | - Chunsheng Liu
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, PR China
| |
Collapse
|
11
|
Sanz J, Randolph HE, Barreiro LB. Genetic and evolutionary determinants of human population variation in immune responses. Curr Opin Genet Dev 2018; 53:28-35. [PMID: 29960896 DOI: 10.1016/j.gde.2018.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 12/18/2022]
Abstract
Humans display remarkable immune response variation when exposed to identical immune challenges. However, our understanding of the genetic, evolutionary, and environmental factors that impact this inter-individual and inter-population immune response heterogeneity is still in its early days. In this review, we discuss three fundamental questions concerning the recent evolution of the human immune system: the degree to which individuals from different populations vary in their innate immune responses, the genetic variants accounting for such differences, and the evolutionary mechanisms that led to the establishment of these variants in modern human populations. We also discuss how past selective events might have contributed to the uneven distribution of immune-related disorders across populations.
Collapse
Affiliation(s)
- Joaquin Sanz
- Department of Biochemistry, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada; Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Haley E Randolph
- Department of Biochemistry, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada; Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Luis B Barreiro
- Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1C5, Canada.
| |
Collapse
|
12
|
Rathnasamy G, Foulds WS, Ling EA, Kaur C. Retinal microglia - A key player in healthy and diseased retina. Prog Neurobiol 2018; 173:18-40. [PMID: 29864456 DOI: 10.1016/j.pneurobio.2018.05.006] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 03/09/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023]
Abstract
Microglia, the resident immune cells of the brain and retina, are constantly engaged in the surveillance of their surrounding neural tissue. During embryonic development they infiltrate the retinal tissues and participate in the phagocytosis of redundant neurons. The contribution of microglia in maintaining the purposeful and functional histo-architecture of the adult retina is indispensable. Within the retinal microenvironment, robust microglial activation is elicited by subtle changes caused by extrinsic and intrinsic factors. When there is a disturbance in the cell-cell communication between microglia and other retinal cells, for example in retinal injury, the activated microglia can manifest actions that can be detrimental. This is evidenced by activated microglia secreting inflammatory mediators that can further aggravate the retinal injury. Microglial activation as a harbinger of a variety of retinal diseases is well documented by many studies. In addition, a change in the microglial phenotype which may be associated with aging, may predispose the retina to age-related diseases. In light of the above, the focus of this review is to highlight the role played by microglia in the healthy and diseased retina, based on findings of our own work and from that of others.
Collapse
Affiliation(s)
- Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore; Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53706, United States
| | - Wallace S Foulds
- Singapore Eye Research Institute Level 6, The Academia, Discovery Tower, 20 College Road, 169856, Singapore; University of Glasgow, Glasgow, Scotland, G12 8QQ, United Kingdom
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, 117594, Singapore.
| |
Collapse
|
13
|
Quickly Attainable and Highly Engrafting Hematopoietic Stem Cells. BLOOD SCIENCE 2018. [DOI: 10.2478/bls-2018-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
14
|
Schaller TH, Batich KA, Suryadevara CM, Desai R, Sampson JH. Chemokines as adjuvants for immunotherapy: implications for immune activation with CCL3. Expert Rev Clin Immunol 2017; 13:1049-1060. [PMID: 28965431 DOI: 10.1080/1744666x.2017.1384313] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Immunotherapy embodies any approach that manipulates the immune system for therapeutic benefit. In this regard, various clinical trials have employed direct vaccination with patient-specific dendritic cells or adoptive T cell therapy to target highly aggressive tumors. Both modalities have demonstrated great specificity, an advantage that is unmatched by other treatment strategies. However, their full potential has yet to be realized. Areas covered: In this review, we provide an overview of chemokines in pathogen and anti-tumor immune responses and discuss further improving immunotherapies by arming particular chemokine axes. Expert commentary: The chemokine macrophage inflammatory protein-1 alpha (MIP-1α, CCL3) has emerged as a potent activator of both innate and adaptive responses. Specifically, CCL3 plays a critical role in recruiting distinct immune phenotypes to intratumoral sites, is a pivotal player in regulating lymph node homing of dendritic cell subsets, and induces antigen-specific T cell responses. The recent breadth of literature outlines the various interactions of CCL3 with these cellular subsets, which have now served as a basis for immunotherapeutic translation.
Collapse
Affiliation(s)
- Teilo H Schaller
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Kristen A Batich
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Carter M Suryadevara
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Rupen Desai
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA
| | - John H Sampson
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b Department of Pathology , Duke University Medical Center , Durham , NC , USA.,c Department of Radiation Oncology , Duke University Medical Center , Durham , NC , USA.,d Department of Immunology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
15
|
Mukaida N, Tanabe Y, Baba T. Chemokines as a Conductor of Bone Marrow Microenvironment in Chronic Myeloid Leukemia. Int J Mol Sci 2017; 18:1824. [PMID: 28829353 PMCID: PMC5578209 DOI: 10.3390/ijms18081824] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/19/2017] [Accepted: 08/20/2017] [Indexed: 12/11/2022] Open
Abstract
All blood lineage cells are generated from hematopoietic stem cells (HSCs), which reside in bone marrow after birth. HSCs self-renew, proliferate, and differentiate into mature progeny under the control of local microenvironments including hematopoietic niche, which can deliver regulatory signals in the form of bound or secreted molecules and from physical cues such as oxygen tension and shear stress. Among these mediators, accumulating evidence indicates the potential involvement of several chemokines, particularly CXCL12, in the interaction between HSCs and bone marrow microenvironments. Fusion between breakpoint cluster region (BCR) and Abelson murine leukemia viral oncogene homolog (ABL)-1 gene gives rise to BCR-ABL protein with a constitutive tyrosine kinase activity and transforms HSCs and/or hematopoietic progenitor cells (HPCs) into disease-propagating leukemia stem cells (LSCs) in chronic myeloid leukemia (CML). LSCs can self-renew, proliferate, and differentiate under the influence of the signals delivered by bone marrow microenvironments including niche, as HSCs can. Thus, the interaction with bone marrow microenvironments is indispensable for the initiation, maintenance, and progression of CML. Moreover, the crosstalk between LSCs and bone marrow microenvironments can contribute to some instances of therapeutic resistance. Furthermore, evidence is accumulating to indicate the important roles of bone marrow microenvironment-derived chemokines. Hence, we will herein discuss the roles of chemokines in CML with a focus on bone marrow microenvironments.
Collapse
MESH Headings
- Animals
- Bone Marrow/metabolism
- Chemokines/metabolism
- Hematopoiesis/genetics
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplastic Stem Cells/metabolism
- Protein Binding
- Receptors, Chemokine/metabolism
- Signal Transduction
- Stem Cell Niche
- Tumor Microenvironment
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Ishikawa, Kanazawa 920-1192, Japan.
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Ishikawa, Kanazawa 920-1192, Japan.
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Ishikawa, Kanazawa 920-1192, Japan.
| |
Collapse
|
16
|
Metzemaekers M, Van Damme J, Mortier A, Proost P. Regulation of Chemokine Activity - A Focus on the Role of Dipeptidyl Peptidase IV/CD26. Front Immunol 2016; 7:483. [PMID: 27891127 PMCID: PMC5104965 DOI: 10.3389/fimmu.2016.00483] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
Chemokines are small, chemotactic proteins that play a crucial role in leukocyte migration and are, therefore, essential for proper functioning of the immune system. Chemokines exert their chemotactic effect by activation of chemokine receptors, which are G protein-coupled receptors (GPCRs), and interaction with glycosaminoglycans (GAGs). Furthermore, the exact chemokine function is modulated at the level of posttranslational modifications. Among the different types of posttranslational modifications that were found to occur in vitro and in vivo, i.e., proteolysis, citrullination, glycosylation, and nitration, NH2-terminal proteolysis of chemokines has been described most intensively. Since the NH2-terminal chemokine domain mediates receptor interaction, NH2-terminal modification by limited proteolysis or amino acid side chain modification can drastically affect their biological activity. An enzyme that has been shown to provoke NH2-terminal proteolysis of various chemokines is dipeptidyl peptidase IV or CD26. This multifunctional protein is a serine protease that preferably cleaves dipeptides from the NH2-terminal region of peptides and proteins with a proline or alanine residue in the penultimate position. Various chemokines possess such a proline or alanine residue, and CD26-truncated forms of these chemokines have been identified in cell culture supernatant as well as in body fluids. The effects of CD26-mediated proteolysis in the context of chemokines turned out to be highly complex. Depending on the chemokine ligand, loss of these two NH2-terminal amino acids can result in either an increased or a decreased biological activity, enhanced receptor specificity, inactivation of the chemokine ligand, or generation of receptor antagonists. Since chemokines direct leukocyte migration in homeostatic as well as pathophysiologic conditions, CD26-mediated proteolytic processing of these chemotactic proteins may have significant consequences for appropriate functioning of the immune system. After introducing the chemokine family together with the GPCRs and GAGs, as main interaction partners of chemokines, and discussing the different forms of posttranslational modifications, this review will focus on the intriguing relationship of chemokines with the serine protease CD26.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Anneleen Mortier
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| |
Collapse
|
17
|
Broxmeyer HE, Capitano M, Campbell TB, Hangoc G, Cooper S. Modulation of Hematopoietic Chemokine Effects In Vitro and In Vivo by DPP-4/CD26. Stem Cells Dev 2016; 25:575-85. [PMID: 26943017 DOI: 10.1089/scd.2016.0026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Dipeptidyl peptidase 4 (DPP4)/CD26 truncates certain proteins, and this posttranslational modification can influence their activity. Truncated (T) colony-stimulating factors (CSFs) are decreased in potency for stimulating proliferation of hematopoietic progenitor cells (HPCs). T-CXCL12, a modified chemokine, is inactive as an HPC chemotactic, survival, and enhancing factor for replating or ex-vivo expansion of HPCs. Moreover, T-CSFs and T-CXCL12 specifically downmodulates the positively acting effects of their own full-length molecule. Other chemokines have DPP4 truncation sites. In the present study, we evaluated effects of DPP4 inhibition (by Diprotin A) or gene deletion of HPC on chemokine inhibition of multicytokine-stimulated HPC, and on chemokine-enhancing effects on single CSF-stimulated HPC proliferation, as well as effects of DPP4 treatment of a number of chemokines. Myelosuppressive effects of chemokines with, but not without, a DPP4 truncation site were greatly enhanced in inhibitory potency by pretreating target bone marrow (BM) cells with Diprotin A, or by assaying their activity on dpp4/cd26(-/-) BM cells. DPP4 treatment of myelosuppressive chemokines containing a DPP4 truncation site produced a nonmyelosuppressive molecule, but one which had the capacity to block suppression by that unmodified chemokine both in vitro and in vivo. Additionally, DPP4 treatment ablated the single cytokine-stimulated HPC-enhancing activity of CCL3/MIP-1α and CCL4/MIP-1β, and blocked the enhancing activity of each unmodified molecule, in vitro and in vivo. These results highlight the functional posttranslational modulating effects of DPP4 on chemokine activities, and information offering additional biological insight into chemokine regulation of hematopoiesis.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Maegan Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Timothy B Campbell
- Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Giao Hangoc
- Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
18
|
Rutar M, Natoli R, Chia RX, Valter K, Provis JM. Chemokine-mediated inflammation in the degenerating retina is coordinated by Müller cells, activated microglia, and retinal pigment epithelium. J Neuroinflammation 2015; 12:8. [PMID: 25595590 PMCID: PMC4308937 DOI: 10.1186/s12974-014-0224-1] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/18/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Monocyte infiltration is involved in the pathogenesis of many retinal degenerative conditions. This process traditionally depends on local expression of chemokines, though the roles of many of these in the degenerating retina are unclear. Here, we investigate expression and in situ localization of the broad chemokine response in a light-induced model of retinal degeneration. METHODS Sprague-Dawley (SD) rats were exposed to 1,000 lux light damage (LD) for up to 24 hrs. At time points during (1 to 24 hrs) and following (3 and 7 days) exposure, animals were euthanized and retinas processed. Microarray analysis assessed differential expression of chemokines. Some genes were further investigated using polymerase chain reaction (PCR) and in situ hybridization and contrasted with photoreceptor apoptosis using terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Recruitment of retinal CD45 (+) leukocytes was determined via fluorescence activated cell sorting (FACS), and expression of chemokine receptors determined using PCR. RESULTS Exposure to 24 hrs of LD resulted in differential expression of chemokines including Ccl3, Ccl4, Ccl7, Cxcl1, and Cxcl10. Their upregulation correlated strongly with peak photoreceptor death, at 24 hrs exposure. In situ hybridization revealed that the modulated chemokines were expressed by a combination of Müller cells, activated microglia, and retinal pigment epithelium (RPE). This preceded large increases in the number of CD45(+) cells at 3- and 7-days post exposure, which expressed a corresponding repertoire of chemokine receptors. CONCLUSIONS Our data indicate that retinal degeneration induces upregulation of a broad chemokine response whose expression is coordinated by Müller cells, microglia, and RPE. The findings inform our understanding of the processes govern the trafficking of leukocytes, which are contributors in the pathology of retinal degenerations.
Collapse
Affiliation(s)
- Matt Rutar
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - Riccardo Natoli
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - R X Chia
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia.
| | - Krisztina Valter
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| | - Jan M Provis
- John Curtin School of Medical Research, The Australian National University, Building 131, Garran Road, Canberra, ACT 2601, Australia. .,ANU Medical School, The Australian National University, 54 Mills Road, Canberra, ACT 2601, Australia.
| |
Collapse
|
19
|
Richter R, Rüster B, Bistrian R, Forssmann WG, Seifried E, Henschler R. Beta-Chemokine CCL15 Affects the Adhesion and Migration of Hematopoietic Progenitor Cells. Transfus Med Hemother 2014; 42:29-37. [PMID: 25960713 DOI: 10.1159/000370168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/27/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Hematopoietic stem and progenitor cell (HPC) motility is essential for HPC transplantation. The chemokine CXCL12 is key for HPC motility. Further regulators are of interest to improve HPC transplantation and regenerative medicine. Here the impact of the human chemokine CCL15 on HPC motility was investigated. METHODS CCL15 plasma concentrations were determined during HPC mobilization in humans. Activity of CCL15 on HPCs was investigated in murine assays, including chemotaxis, adhesion, and CFU-A assays, and competitive repopulation assays. RESULTS During HPC mobilization with granulocyte colony-stimulating factor, blood plasma contains increased concentrations (1.1 ± 0.1 ng/ml) of activated CCL15(27-92) versus 0.4 ± 0.1 ng/ml in controls (p = 0.02). CCL15(27-92) significantly enhanced CXCL12-induced transwell migration of Lin-/Sca1+ HPCs and strengthened shear stress-dependent adhesion to vascular cell adhesion molecule-1 (VCAM-1). CCL15(27-92) dose-dependently reduced the colony size in CFU-A assays performed with murine bone marrow and Lin-/Sca1+ HPCs. CCL15(27-92) did not show a direct impact on cell cycle status of HPCs. In murine repopulation assays, pretreatment of bone marrow with CCL15(27-92) significantly increased competitive repopulation. CONCLUSION Our results point to a regulation of HPCs by CCL15 by modulating migratory and adhesive properties of HPCs with the potency to improve HPC short-term engraftment in stem cell transplantation.
Collapse
Affiliation(s)
- Rudolf Richter
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt/M., Germany ; Clinic of Immunology and Rheumatology, Hannover Medical School, Hanover, Germany
| | - Brigitte Rüster
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt/M., Germany
| | - Roxana Bistrian
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt/M., Germany
| | - Wolf-Georg Forssmann
- Clinic of Immunology and Rheumatology, Hannover Medical School, Hanover, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt/M., Germany
| | - Reinhard Henschler
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt/M., Germany
| |
Collapse
|
20
|
Fulkerson PC, Schollaert KL, Bouffi C, Rothenberg ME. IL-5 triggers a cooperative cytokine network that promotes eosinophil precursor maturation. THE JOURNAL OF IMMUNOLOGY 2014; 193:4043-52. [PMID: 25230753 DOI: 10.4049/jimmunol.1400732] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Eosinophils originate in the bone marrow from an eosinophil lineage-committed, IL-5Rα-positive, hematopoietic progenitor (eosinophil progenitor). Indeed, IL-5 is recognized as a critical regulator of eosinophilia and has effects on eosinophil progenitors, eosinophil precursors, and mature eosinophils. However, substantial levels of eosinophils remain after IL-5 neutralization or genetic deletion, suggesting that there are alternative pathways for promoting eosinophilia. In this study, we investigated the contributory role of IL-5 accessory cytokines on the final stages of eosinophil differentiation. IL-5 stimulation of low-density bone marrow cells resulted in expression of a panel of cytokines and cytokine receptors, including several ligand-receptor pairs. Notably, IL-4 and IL-4Rα were expressed by eosinophil precursors and mature eosinophils. Signaling through IL-4Rα promoted eosinophil maturation when IL-5 was present, but IL-4 stimulation in the absence of IL-5 resulted in impaired eosinophil survival, suggesting that IL-4 cooperates with IL-5 to promote eosinophil differentiation. In contrast, CCL3, an eosinophil precursor-produced chemokine that signals through CCR1, promotes terminal differentiation of CCR1-positive eosinophil precursors in the absence of IL-5, highlighting an autocrine loop capable of sustaining eosinophil differentiation. These findings suggest that brief exposure to IL-5 is sufficient to initiate a cytokine cooperative network that promotes eosinophil differentiation of low-density bone marrow cells independent of further IL-5 stimulation.
Collapse
Affiliation(s)
- Patricia C Fulkerson
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Kaila L Schollaert
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Carine Bouffi
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
21
|
Baba T, Mukaida N. Role of macrophage inflammatory protein (MIP)-1α/CCL3 in leukemogenesis. Mol Cell Oncol 2014; 1:e29899. [PMID: 27308309 PMCID: PMC4905173 DOI: 10.4161/mco.29899] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/27/2022]
Abstract
The biologic function of the CC chemokine macrophage inflammatory protein-1α (MIP-1α/CCL3) has been extensively studied since its initial identification as a macrophage-derived inflammatory mediator. In addition to its proinflammatory activities, CCL3 negatively regulates the proliferation of hematopoietic stem/progenitor cells (HSPCs). On the basis of this unique function, CCL3 is alternatively referred to as a stem cell inhibitor. This property has prompted many researchers to investigate the effects of CCL3 on normal physiologic hematopoiesis and pathophysiologic processes of hematopoietic malignancies. Consequently, there is accumulating evidence supporting a crucial involvement of CCL3 in the pathophysiology of several types of leukemia arising from neoplastic transformation of HSPCs. In this review we discuss the roles of CCL3 in leukemogenesis and its potential value as a target in a novel therapeutic strategy for the treatment of leukemia.
Collapse
Affiliation(s)
- Tomohisa Baba
- Division of Molecular Bioregulation; Cancer Research Institute; Kanazawa University; Kanazawa, Ishikawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation; Cancer Research Institute; Kanazawa University; Kanazawa, Ishikawa, Japan
| |
Collapse
|
22
|
Baba T, Naka K, Morishita S, Komatsu N, Hirao A, Mukaida N. MIP-1α/CCL3-mediated maintenance of leukemia-initiating cells in the initiation process of chronic myeloid leukemia. J Exp Med 2013; 210:2661-2673. [PMID: 24166712 PMCID: PMC3832924 DOI: 10.1084/jem.20130112] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 09/26/2013] [Indexed: 12/13/2022] Open
Abstract
In the initiation process of chronic myeloid leukemia (CML), a small number of transformed leukemia-initiating cells (LICs) coexist with a large number of normal hematopoietic cells, gradually increasing thereafter and eventually predominating in the hematopoietic space. However, the interaction between LICs and normal hematopoietic cells at the early phase has not been clearly delineated because of the lack of a suitable experimental model. In this study, we succeeded in causing a marked leukocytosis resembling CML from restricted foci of LICs in the normal hematopoietic system by direct transplantation of BCR-ABL gene-transduced LICs into the bone marrow (BM) cavity of nonirradiated mice. Herein, we observed that BCR-ABL(+)lineage(-)c-kit(-) immature leukemia cells produced high levels of an inflammatory chemokine, MIP-1α/CCL3, which promoted the development of CML. Conversely, ablation of the CCL3 gene in LICs dramatically inhibited the development of CML and concomitantly reduced recurrence after the cessation of a short-term tyrosine kinase inhibitor treatment. Finally, normal hematopoietic stem/progenitor cells can directly impede the maintenance of LICs in BM in the absence of CCL3 signal.
Collapse
MESH Headings
- Animals
- Bone Marrow Transplantation
- Chemokine CCL3/deficiency
- Chemokine CCL3/genetics
- Chemokine CCL3/metabolism
- Disease Models, Animal
- Genes, abl
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Nude
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/transplantation
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Proto-Oncogene Proteins c-kit/metabolism
- Receptors, CCR1/deficiency
- Receptors, CCR1/genetics
- Receptors, CCR1/metabolism
- Receptors, CCR5/deficiency
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Tomohisa Baba
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Kazuhito Naka
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation and Department of Hematology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Norio Komatsu
- Department of Transfusion Medicine and Stem Cell Regulation and Department of Hematology, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Atsushi Hirao
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Exploratory Project on Cancer Stem Cells, and Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa 920-1192, Ishikawa, Japan
| |
Collapse
|
23
|
Chitu V, Nacu V, Charles JF, Henne WM, McMahon HT, Nandi S, Ketchum H, Harris R, Nakamura MC, Stanley ER. PSTPIP2 deficiency in mice causes osteopenia and increased differentiation of multipotent myeloid precursors into osteoclasts. Blood 2012; 120:3126-35. [PMID: 22923495 PMCID: PMC3471520 DOI: 10.1182/blood-2012-04-425595] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/07/2012] [Indexed: 12/11/2022] Open
Abstract
Missense mutations that reduce or abrogate myeloid cell expression of the F-BAR domain protein, proline serine threonine phosphatase-interacting protein 2 (PSTPIP2), lead to autoinflammatory disease involving extramedullary hematopoiesis, skin and bone lesions. However, little is known about how PSTPIP2 regulates osteoclast development. Here we examined how PSTPIP2 deficiency causes osteopenia and bone lesions, using the mouse PSTPIP2 mutations, cmo, which fails to express PSTPIP2 and Lupo, in which PSTPIP2 is dysfunctional. In both models, serum levels of the pro-osteoclastogenic factor, MIP-1α, were elevated and CSF-1 receptor (CSF-1R)-dependent production of MIP-1α by macrophages was increased. Treatment of cmo mice with a dual specificity CSF-1R and c-Kit inhibitor, PLX3397, decreased circulating MIP-1α and ameliorated the extramedullary hematopoiesis, inflammation, and osteopenia, demonstrating that aberrant myelopoiesis drives disease. Purified osteoclast precursors from PSTPIP2-deficient mice exhibit increased osteoclastogenesis in vitro and were used to probe the structural requirements for PSTPIP2 suppression of osteoclast development. PSTPIP2 tyrosine phosphorylation and a functional F-BAR domain were essential for PSTPIP2 inhibition of TRAP expression and osteoclast precursor fusion, whereas interaction with PEST-type phosphatases was only required for suppression of TRAP expression. Thus, PSTPIP2 acts as a negative feedback regulator of CSF-1R signaling to suppress inflammation and osteoclastogenesis.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
INTRODUCTION Pharmacological mobilization has been exploited as a means to obtain hematopoietic stem progenitor cells (HSPCs) for hematopoietic reconstitution. HSPCs mobilized from bone marrow into peripheral blood (PB) are a preferred source of stem cells for transplantation, because they are easily accessible and evidence indicates that they engraft faster after transplantation than HSPCs directly harvested from bone marrow (BM) or umbilical cord blood (UCB). AREAS COVERED Since chemokine-chemokine receptor axes are involved in retention of HSPCs in the BM microenvironment, chemokine receptor agonists have been proposed as therapeutics to facilitate the mobilization process. These compounds include agonists of the CXCR4 receptor expressed on HSPCs (CTCE-0021 and ATI-2341) or chemokines binding to chemokine receptors expressed on granuclocytes and monocytes (e.g., CXCL2, also known as the growth-related oncogene protein-beta (Gro-β); CCL3, also known as macrophage inflammatory protein-1α (MIP-1α); or CXCL8, also known as IL-8) could be employed alone or in combination with other mobilizing agents (e.g., G-CSF or Plerixafor (AMD3100)). We discuss the current state of knowledge about chemokine receptor agonists and the rationale for their application in mobilization protocols. EXPERT OPINION Evidence is accumulating that CXCR4 receptor agonists could be employed alone or with other agents as mobilizing drugs. In particular they may provide an alternative for patients that are poor mobilizers.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- University of Louisville, Stem Cell Institute at James Graham Brown Cancer Center, 500 S. Floyd Street, Room. 107, Louisville, KY 40202, USA.
| | | |
Collapse
|
25
|
Abstract
Monocytes originate from progenitors in the bone marrow and traffic via the bloodstream to peripheral tissues. During both homeostasis and inflammation, circulating monocytes leave the bloodstream and migrate into tissues where, following conditioning by local growth factors, pro-inflammatory cytokines and microbial products, they differentiate into macrophage or dendritic cell populations. Recruitment of monocytes is essential for effective control and clearance of viral, bacterial, fungal and protozoal infections, but recruited monocytes also contribute to the pathogenesis of inflammatory and degenerative diseases. The mechanisms that control monocyte trafficking under homeostatic, infectious and inflammatory conditions are being unravelled and are the focus of this Review.
Collapse
|
26
|
Ortega MT, Xie L, Mora S, Chapes SK. Evaluation of macrophage plasticity in brown and white adipose tissue. Cell Immunol 2011; 271:124-33. [PMID: 21757190 PMCID: PMC3168070 DOI: 10.1016/j.cellimm.2011.06.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 06/09/2011] [Accepted: 06/13/2011] [Indexed: 01/02/2023]
Abstract
There are still questions about whether macrophage differentiation is predetermined or is induced in response to tissue microenvironments. C2D macrophage cells reside early in the macrophage lineage in vitro, but differentiate to a more mature phenotype after adoptive transfer to the peritoneal cavity (PEC-C2D). Since C2D macrophage cells also traffic to adipose tissue after adoptive transfer, we explored the impact of white adipose tissue (WAT), brown adipose tissue (BAT) and in vitro cultured adipocytes on C2D macrophage cells. When PEC-C2D macrophage cells were cultured with preadipocytes the cells stretched out and CD11b and Mac-2 expression was lower compared to PEC-C2D macrophage cells placed in vitro alone. In contrast, PEC-C2D cells co-cultured with adipocytes maintained smaller, round morphology and more cells expressed Mac-2 compared to PEC-C2D co-cultured with preadipocytes. After intraperitoneal injection, C2D macrophage cells migrated into both WAT and BAT. A higher percentage of C2D macrophage cells isolated from WAT (WAT-C2D) expressed Ly-6C (33%), CD11b (11%), Mac-2 (11%) and F4/80 (29%) compared to C2D macrophage cells isolated from BAT (BAT-C2D). Overall, BAT-C2D macrophage cells had reduced expression of many cytokine, chemokine and receptor gene transcripts when compared to in vitro grown C2D macrophages, while WAT-C2D macrophage cells and PEC-C2D up-regulated many of these gene transcripts. These data suggest that the C2D macrophage phenotype can change rapidly and distinct phenotypes are induced by different microenvironments.
Collapse
Affiliation(s)
- M. Teresa Ortega
- Division of Biology, Kansas State University, Manhattan, KS, 66506
| | - Linglin Xie
- Division of Biology, Kansas State University, Manhattan, KS, 66506
| | - Silvia Mora
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, The University of Liverpool, Crown Street, Liverpool L69 3BX, United Kingdom
| | | |
Collapse
|
27
|
|
28
|
Kränkel N, Spinetti G, Amadesi S, Madeddu P. Targeting stem cell niches and trafficking for cardiovascular therapy. Pharmacol Ther 2011; 129:62-81. [PMID: 20965213 PMCID: PMC3017934 DOI: 10.1016/j.pharmthera.2010.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/06/2010] [Indexed: 12/12/2022]
Abstract
Regenerative cardiovascular medicine is the frontline of 21st-century health care. Cell therapy trials using bone marrow progenitor cells documented that the approach is feasible, safe and potentially beneficial in patients with ischemic disease. However, cardiovascular prevention and rehabilitation strategies should aim to conserve the pristine healing capacity of a healthy organism as well as reactivate it under disease conditions. This requires an increased understanding of stem cell microenvironment and trafficking mechanisms. Engagement and disengagement of stem cells of the osteoblastic niche is a dynamic process, finely tuned to allow low amounts of cells move out of the bone marrow and into the circulation on a regular basis. The balance is altered under stress situations, like tissue injury or ischemia, leading to remarkably increased cell egression. Individual populations of circulating progenitor cells could give rise to mature tissue cells (e.g. endothelial cells or cardiomyocytes), while the majority may differentiate to leukocytes, affecting the environment of homing sites in a paracrine way, e.g. promoting endothelial survival, proliferation and function, as well as attenuating or enhancing inflammation. This review focuses on the dynamics of the stem cell niche in healthy and disease conditions and on therapeutic means to direct stem cell/progenitor cell mobilization and recruitment into improved tissue repair.
Collapse
Affiliation(s)
- Nicolle Kränkel
- Institute of Physiology/Cardiovascular Research, University of Zürich, and Cardiovascular Center, Cardiology, University Hospital Zurich, Zürich, Switzerland
| | | | - Silvia Amadesi
- Experimental Cardiovascular Medicine, University of Bristol, Bristol, UK
| | - Paolo Madeddu
- Experimental Cardiovascular Medicine, University of Bristol, Bristol, UK
| |
Collapse
|
29
|
|
30
|
CCR1 knockdown suppresses human non-small cell lung cancer cell invasion. J Cancer Res Clin Oncol 2008; 135:695-701. [PMID: 18972130 DOI: 10.1007/s00432-008-0505-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Accepted: 10/10/2008] [Indexed: 01/02/2023]
Abstract
PURPOSE CC chemokine receptor 1 (CCR1) plays a critical role in the recruitment of leukocytes to the site of inflammation. Tumor invasion and metastasis share many similarities with leukocyte trafficking, which is critically regulated by chemokines and their receptors. In this study, we aimed to assess the role of CCR1 in non-small cell lung cancer (NSCLC). METHODS CCR1 expression was determined by Western blotting in two human NSCLC clones (95C and 95D) with different metastatic potential. We silenced CCR1 expression through microRNA-mediated RNA interference, and examined the invasiveness and proliferation of CCR1-silenced NSCLC cell through Matrigel assay and MTT assay. Matrix metalloproteinases (MMPs) activity was determined by gelatin zymography. RESULTS We found that expression of CCR1 was correlated with the aggressive phenotype of the NSCLC cells. CCR1 knockdown significantly suppressed the invasiveness of NSCLC cells, but had only a minor effect on cell proliferation. Moreover, we demonstrated that CCR1 knockdown significantly reduced the expression level of matrix metalloproteinase-9. CONCLUSIONS These findings suggest that CCR1 contributes to NSCLC cell migration by stimulating cell invasion, independent of cell proliferation. CCR1 might be a new target for NSCLC therapy.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Granulocyte colony-stimulating factor-mobilized peripheral blood stem cells are widely used to reconstitute hematopoiesis; however, preclinical and clinical studies show that improvements to this mobilization can be achieved. We discuss the development of new mobilizing regimens and evaluation of new findings on mobilized stem cell populations that may improve the utility and convenience of peripheral blood stem cell transplant. RECENT FINDINGS Chemokines and their receptors regulate leukocyte trafficking, and altering chemokine signaling pathways mobilizes stem cells. In recent trials, combination use of the chemokine (C-X-C motif) receptor 4 antagonist AMD3100 and granulocyte colony-stimulating factor mobilized more CD34 cells in fewer days than granulocyte colony-stimulating factor alone and allowed more patients to proceed to autotransplant. In preclinical studies the chemokine GRObeta synergizes with granulocyte colony-stimulating factor and when used alone or with granulocyte colony-stimulating factor mobilizes more primitive hematopoietic stem cells with less apoptosis, higher integrin activation, lower CD26 expression and enhanced marrow homing compared with granulocyte colony-stimulating factor. Hematopoietic stem cells mobilized by GRObeta or AMD3100 demonstrate superior engraftment and contribution to chimerism in primary and secondary transplant studies in mice, and peripheral blood stem cells mobilized by AMD3100 and granulocyte colony-stimulating factor in patients demonstrate enhanced engraftment capabilities in immunodeficient mice. SUMMARY Alternate regimens differentially mobilize stem cell populations with unique intrinsic properties with the potential to expand the utility of hematopoietic transplantation. Continued mechanistic evaluation will be critical to our understanding of mechanisms of mobilization and their use in regenerative medicine.
Collapse
Affiliation(s)
- Louis M Pelus
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| |
Collapse
|
32
|
Pelus LM, Fukuda S. Chemokine-mobilized adult stem cells; defining a better hematopoietic graft. Leukemia 2008; 22:466-73. [PMID: 17972941 PMCID: PMC2814589 DOI: 10.1038/sj.leu.2405021] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 10/04/2007] [Accepted: 10/08/2007] [Indexed: 12/22/2022]
Abstract
Stem cell research is currently focused on totipotent stem cells and their therapeutic potential, however adult stem cells, while restricted to differentiation within their tissue or origin, also have therapeutic utility. Transplantation with bone marrow hematopoietic stem cells (HSC) has been used for curative therapy for decades. More recently, alternative sources of HSC, particularly those induced to exit marrow or mobilize to peripheral blood by G-CSF, have become the most widely used hematopoietic graft and show significant superiority to marrow HSC. The chemokine/chemokine receptor axis also mobilizes HSC that occurs more rapidly than with G-CSF. In mice, the HSC and progenitor cells (HPC) mobilized by the CXCR2 receptor agonist GRObeta can be harvested within minutes of administration and show significantly lower levels of apoptosis, enhanced homing to marrow, expression of more activated integrin receptors and superior repopulation kinetics and more competitive engraftment than the equivalent cells mobilized by G-CSF. These characteristics suggest that chemokine axis-mobilized HSC represent a population of adult stem cells distinct from those mobilized by G-CSF, with superior therapeutic potential. It remains to be determined if the chemokine mobilization axis can be harnessed to mobilize other populations of unique adult stem cells with clinical utility.
Collapse
Affiliation(s)
- L M Pelus
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | |
Collapse
|
33
|
Abstract
Herein we have analyzed chemokine involvement in the trafficking of developing and mature mouse natural killer (NK) cells in the bone marrow (BM). We observed drastic changes of CCR1, CXCR3, and CXCR4 expression and function during progression from precursor NK (pNK) cells to immature DX5- NK (iNK) and mature DX5+ NK (mNK) cells. pNK and mNK cells expressed the 3 receptors, while only CXCR4 was detected on iNK cells. Correspondingly, mNK cells migrated to CXCL12, CXCL10, and CCL3, and pNK and iNK cells to CXCL12, whereas pNK cells migrated to CCL3 and CXCL10 only after CXCL12 stimulation. Comparison of BM, peripheral blood, and spleen mNK cell populations revealed that CXCL12, CXCL10, and CCL3 preferentially affected BM mNK cell migration. Administration of the CXCR4 antagonist, AMD-3100, to C57BL/6 mice induced strong reduction of mNK and iNK cells in the BM and increased their number in blood and spleen. Conversely, CCL3 administration selectively mobilized mNK cells from the BM and this effect correlated with its ability to inhibit CXCL12-mediated mNK cell responses in vitro. Our results suggest that the combined action of chemokines selectively regulates localization of NK cell subsets in the BM and direct their maturation and migration to the periphery.
Collapse
|
34
|
Lee JE, Shin HH, Lee EA, Van Phan T, Choi HS. Stimulation of osteoclastogenesis by enhanced levels of MIP-1alpha in BALB/c mice in vitro. Exp Hematol 2007; 35:1100-8. [PMID: 17588479 DOI: 10.1016/j.exphem.2007.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 03/14/2007] [Accepted: 04/09/2007] [Indexed: 11/23/2022]
Abstract
OBJECTIVES We compared osteoclast (OC) formation in bone marrow-derived macrophages (BMM) from C57BL/6 (B/6) and BALB/c (B/c) mice. After stimulation of receptor activator of nuclear factor-kappaB ligand (RANKL), enhanced OC formation and higher level of macrophage inflammatory protein-1alpha (MIP-1alpha) were observed in the BMM from B/c mice. In this study, we determined whether MIP-1alpha is responsible for stimulated OC formation in the BMM. MATERIALS AND METHODS OC formation was evaluated in BMM. Expression of MIP-1alpha during OC formation was analyzed at the mRNA and protein levels. Apoptosis of mature OCs was evaluated by observing the degradation of DNA. Activation of nuclear factor-kappaB (NF-kappaB) was measured by electrophoretic mobility shift assay. RESULTS After stimulation by RANKL expression of MIP-1alpha at the mRNA and protein levels was much higher in BMM from B/c mice than in BMM from B/6 mice. Transcripts of the MIP-1alpha receptors, CCR1 and CCR5, were present at similar levels in unstimulated BMM of the two strains. Blockade of MIP-1alpha inhibited OC formation, and exogenously added MIP-1alpha stimulated it in RANKL-stimulated BMM. MIP-1alpha affected not only the early precursors but also mature OCs. It prevented apoptosis of mature OCs by activating NF-kappaB, and the effect of RANKL on survival was dependent on its ability to induce MIP-1alpha. CONCLUSIONS MIP-1alpha, induced by RANKL during OC differentiation, increases OC formation by acting on OC progenitor cells, and prolongs survival of mature OC via signaling through NF-kappaB. The enhanced OC formation in BMM from B/c mice could be due to, at least in part, to their higher levels of MIP-1alpha.
Collapse
Affiliation(s)
- Ji-Eun Lee
- Department of Biological Sciences and Immunomodulation Research Center, University of Ulsan, Ulsan, Korea
| | | | | | | | | |
Collapse
|
35
|
Broxmeyer HE, Sehra S, Cooper S, Toney LM, Kusam S, Aloor JJ, Marchal CC, Dinauer MC, Dent AL. Aberrant regulation of hematopoiesis by T cells in BAZF-deficient mice. Mol Cell Biol 2007; 27:5275-85. [PMID: 17526724 PMCID: PMC1952080 DOI: 10.1128/mcb.01967-05] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The BAZF (BCL-6b) protein is highly similar to the BCL-6 transcriptional repressor. While BCL-6 has been characterized extensively, relatively little is known about the normal function of BAZF. In order to understand the physiological role of BAZF, we created BAZF-deficient mice. Unlike BCL-6-deficient mice, BAZF-deficient mice are healthy and normal in size. However, BAZF-deficient mice have a hematopoietic progenitor phenotype that is almost identical to that of BCL-6-deficient mice. Compared to wild-type mice, both BAZF-deficient and BCL-6-deficient mice have greatly reduced numbers of cycling hematopoietic progenitor cells (HPC) in the BM and greatly increased numbers of cycling HPC in the spleen. In contrast to HPC from wild-type mice, HPC from BAZF-deficient and BCL-6-deficient mice are resistant to chemokine-induced myelosuppression and do not show a synergistic growth response to granulocyte-macrophage colony-stimulating factor plus stem cell factor. Depletion of CD8 T cells in BAZF-deficient mice reverses several of the hematopoietic defects in these mice. Since both BAZF- and BCL-6-deficient mice have defects in CD8 T-cell differentiation, we hypothesize that both BCL-6 and BAZF regulate HPC homeostasis by an indirect pathway involving CD8 T cells.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology and The Walther Oncology Center, 950 W. Walnut St. R2 302, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cittera E, Leidi M, Buracchi C, Pasqualini F, Sozzani S, Vecchi A, Waterfield JD, Introna M, Golay J. The CCL3 Family of Chemokines and Innate Immunity Cooperate In Vivo in the Eradication of an Established Lymphoma Xenograft by Rituximab. THE JOURNAL OF IMMUNOLOGY 2007; 178:6616-23. [PMID: 17475893 DOI: 10.4049/jimmunol.178.10.6616] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The therapeutic mAb rituximab induced the expression of the CCL3 and CCL4 chemokines in the human lymphoma line BJAB following binding to the CD20 Ag. Induction of CCL3/4 in vitro was specific, was observed in several cell lines and freshly isolated lymphoma samples and also took place at the protein level in vitro and in vivo. To investigate the role of these beta-chemokines in the mechanism of action of rituximab, we synthesized a N-terminally truncated CCL3 molecule CCL3(11-70), which had antagonist activity on chemotaxis mediated by either CCL3 or BJAB supernatant. We also set up an established s.c. BJAB tumor model in athymic mice. Rituximab, given weekly after tumors had reached 250 mm2, led to complete disappearance of the lymphoma within 2-3 wk. Treatment of mice with cobra venom factor showed that complement was required for rituximab therapeutic activity. Treatment of BJAB tumor bearing mice every 2 days with the CCL3(11-70) antagonist, starting 1 wk before rituximab treatment, had no effect on tumor growth by itself, but completely inhibited the therapeutic activity of the Ab. To determine whether CCL3 acts through recruitment/activation of immune cells, we specifically depleted NK cells, polymorphonuclear cells, and macrophages using mAbs, clodronate treatment, or Rag2-/-cgamma-/- mice. The data demonstrated that these different cell populations are involved in BJAB tumor eradication. We propose that rituximab rapidly activates complement and induces beta-chemokines in vivo, which in turn activate the innate immunity network required for efficient eradication of the bulky BJAB tumor.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Burkitt Lymphoma/immunology
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/therapy
- Cell Line, Tumor
- Chemokine CCL3
- Chemokine CCL4
- Chemokines, CC/biosynthesis
- Chemokines, CC/genetics
- Chemokines, CC/physiology
- Complement System Proteins/physiology
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Immunity, Innate/genetics
- Male
- Mice
- Mice, Nude
- Multigene Family/genetics
- Multigene Family/immunology
- RNA, Messenger/biosynthesis
- Rituximab
- Transplantation, Heterologous/immunology
Collapse
Affiliation(s)
- Elena Cittera
- Laboratory of Cellular and Gene Therapy G. Lanzani, Division of Haematology, Ospedali Riuniti di Bergamo, Bergamo, and Section of General Pathology, University of Brescia, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Broxmeyer HE, Hangoc G, Cooper S, Campbell T, Ito S, Mantel C. AMD3100 and CD26 modulate mobilization, engraftment, and survival of hematopoietic stem and progenitor cells mediated by the SDF-1/CXCL12-CXCR4 axis. Ann N Y Acad Sci 2007; 1106:1-19. [PMID: 17360804 DOI: 10.1196/annals.1392.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The chemokine stromal cell-derived factor-1 (SDF-1/CXCL12) and its receptor, CXCR4, are involved in a number of facets of the regulation of hematopoiesis at the level of hematopoietic stem (HSCs) and progenitor (HPCs) cells. Modulation of this ligand-receptor interaction may be of clinical utility. We now report that: (1) the CC chemokine, macrophage inflammatory protein-1alpha (MIP-1alpha/CCL3) synergizes with AMD3100 (an antagonist of the binding of SDF-1/CXCL12 to CXCR4) to rapidly mobilize HPCs to the blood of mice; moreover, the combination of granulocyte colony-stimulating factor (G-CSF) with AMD3100 and MIP-1alpha/CCL3, given in a specific sequence, mobilizes the greatest number of HPCs compared to any combination of two of these mobilizing agents; (2) pretreatment of recipient mice with Diprotin A, an inhibitor of CD26/Dipeptidylpeptidase IV (DPPIV), enhances the competitive HSCs repopulating capacity of untreated donor cells; (3) the survival-enhancing effects of SDF-1/CXCL12 on HPCs subjected in vitro to delayed addition of growth factors (GFs) are mediated in part through the cell cycle-related proteins p21(cip1/waf1) (as assessed using p21(cip1/waf1) -/- and +/+ mice) and Mad2 (using Mad2 +/- and +/+ mice); and (4) deletion of CD26/DPPIV on mouse bone marrow cells increases the survival-enhancing effects of SDF-1/CXCL12 on HPCs. These results demonstrate the means to increase the mobilization of HPCs, the engrafting capability of HSCs, and responsiveness of HPCs to the survival-enhancing activity of SDF-1/CXCL12, effects that may be of practical value.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, and the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Franzke A, Geffers R, Hunger JK, Pförtner S, Piao W, Ivanyi P, Grosse J, Probst-Kepper M, Ganser A, Buer J. Identification of novel regulators in T-cell differentiation of aplastic anemia patients. BMC Genomics 2006; 7:263. [PMID: 17052335 PMCID: PMC1626471 DOI: 10.1186/1471-2164-7-263] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 10/19/2006] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Aplastic anemia (AA) is a bone marrow failure syndrome mostly characterized by an immune-mediated destruction of marrow hematopoietic progenitor/stem cells. The resulting hypocellularity limits a detailed analysis of the cellular immune response. To overcome this technical problem we performed a microarray analysis of CD3+ T-cells derived from bone marrow aspirates and peripheral blood samples of newly diagnosed AA patients and healthy volunteers. Two AA patients were additionally analyzed after achieving a partial remission following immunosuppression. The regulation of selected candidate genes was confirmed by real-time RT-PCR. RESULTS Among more than 22,200 transcripts, 583 genes were differentially expressed in the bone marrow of AA patients compared to healthy controls. Dysregulated genes are involved in T-cell mediated cytotoxicity, immune response of Th1 differentiated T-cells, and major regulators of immune function. In hematological remission the expression levels of several candidate genes tend to normalize, such as immune regulators and genes involved in proinflammatory immune response. CONCLUSION Our study suggests a pivotal role of Th1/Tc1 differentiated T-cells in immune-mediated marrow destruction of AA patients. Most importantly, immune regulatory genes could be identified, which are likely involved in the recovery of hematopoiesis and may help to design new therapeutic strategies in bone marrow failure syndromes.
Collapse
Affiliation(s)
- Anke Franzke
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Robert Geffers
- Department of Cell Biology and Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - J Katrin Hunger
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Susanne Pförtner
- Department of Cell Biology and Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D-38124 Braunschweig, Germany
| | - Wenji Piao
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Philipp Ivanyi
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Jens Grosse
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Michael Probst-Kepper
- Junior Research Group for Xenotransplantation, Department of Visceral and Transplant Surgery, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis and Oncology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| | - Jan Buer
- Department of Cell Biology and Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, D-38124 Braunschweig, Germany
- Department of Medical Microbiology, Hannover Medical School, Carl-Neuberg-Str.1, D-30625 Hannover, Germany
| |
Collapse
|
39
|
Broxmeyer HE, Pelus LM, Kim CH, Hangoc G, Cooper S, Hromas R. Synergistic inhibition in vivo of bone marrow myeloid progenitors by myelosuppressive chemokines and chemokine-accelerated recovery of progenitors after treatment of mice with Ara-C. Exp Hematol 2006; 34:1069-77. [PMID: 16863913 DOI: 10.1016/j.exphem.2006.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Selected chemokines suppress proliferation of hematopoietic progenitor cells (HPCs) in vitro; some of these have demonstrated inhibition of myelopoiesis in vivo. Because myelosuppressive chemokines synergize in vitro with other myelosuppressive chemokines, we sought to determine whether additional chemokines active in vitro were myelosuppressive in vivo and whether combinations of myelosuppressive chemokines synergized in vivo to dampen myelopoiesis. We also evaluated three chemokines in vivo for myeloprotection against Ara-C-induced decreases in HPCs. METHODS C3H/HeJ mice were used for analysis of in vivo influence of chemokines, with the end points being effects on absolute numbers and cycling status of HPCs. RESULTS When used alone, CCL2, CCL3, CCL19, CCL20, CXCL4, CXCL5, CXCL8, CXCL9, and XCL1 caused dose-dependent significant decreases in absolute numbers and cycling status of HPCs in vivo. The following combinations of two chemokines resulted in in vivo myelosuppression at concentrations much lower than that induced by each chemokine alone: CCL3 plus either CXCL8 or CXCL4, CXCL8 plus CXCL4, CCL2 plus either CCL20 or CXCL9, CCL20 plus CXCL9, CXCL5 plus either XCL1 or CCL19, XCL1 plus CCL19, and CCL3 plus CCL19. Also, mice injected with CXCL8, CXCL4, or the chimeric CXCL8/CXCL4 protein CXCL8M1 manifested accelerated recovery of absolute numbers of HPCs in response to the toxic effects of Ara-C administration. CONCLUSIONS A number of chemokines shown previously to manifest inhibitory effects in vitro for proliferation of HPCs are now demonstrated to also induce myelosuppression in vivo. Moreover, combinations of low dosages of two myelosuppressive chemokines when administered together demonstrate synergistic suppression in vivo. Additionally, chemokines, including a CXCL8M1 chimeric protein previously shown to manifest enhanced suppression of HPC proliferation in vitro and in vivo, accelerate HPC recovery after treatment of mice with Ara-C. These results may be of use for future clinical utility of chemokines in a myelosuppressive/myeloprotective setting.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Departments of Microbiology and Immunology, and the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Pelus LM, Fukuda S. Peripheral blood stem cell mobilization: the CXCR2 ligand GRObeta rapidly mobilizes hematopoietic stem cells with enhanced engraftment properties. Exp Hematol 2006; 34:1010-20. [PMID: 16863907 DOI: 10.1016/j.exphem.2006.04.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Indexed: 11/29/2022]
Abstract
Chemokines direct the movement of leukocytes, including hematopoietic stem and progenitor cells, and can mobilize hematopoietic cells from marrow to peripheral blood where they can be used for transplantation. In this review, we will discuss the stem cell mobilizing activities and mechanisms of action of GRObeta, a CXC chemokine ligand for the CXCR2 receptor. GRObeta rapidly mobilizes short- and long-term repopulating cells in mice and/or monkeys and synergistically enhances mobilization responses when combined with the widely used clinical mobilizer, granulocyte colony-stimulating factor (G-CSF). The hematopoietic graft mobilized by GRObeta contains significantly more CD34(neg), Sca-1+, c-kit+, lineage(neg) (SKL) cells than the graft mobilized by G-CSF. In mice, stem cells mobilized by GRObeta demonstrate a competitive advantage upon long-term repopulation analysis and restore neutrophil and platelet counts significantly faster than cells mobilized by G-CSF. Even greater advantage in repopulation and restoration of hematopoiesis are observed with stem cells mobilized by the combination of GRObeta and G-CSF. GRObeta-mobilized SKL cells demonstrate enhanced adherence to vascular cell adhesion molecule-1 and VCAM(pos) endothelial cells and home more efficiently to bone marrow in vivo. The marrow homing ability of GRObeta-mobilized cells is less dependent on the CXCR4/SDF-1 axis than cells mobilized by G-CSF. The mechanism of mobilization by GRObeta requires active matrix metalloproteinase-9 (MMP-9), which results from release of pro-MMP-9 from peripheral blood, and marrow neutrophils, which alters the stoichiometry between pro-MMP-9 and its inhibitor tissue inhibitor of metalloproteinase-1, resulting in MMP-9 activation. The efficacy and rapid action of GRObeta and lack of proinflammatory activity make it an attractive agent to supplement mobilization by G-CSF. In addition, GRObeta may also have clinical mobilizing efficacy on its own, reducing the overall time and costs associated with peripheral blood stem cell transplantation.
Collapse
Affiliation(s)
- Louis M Pelus
- Department of Microbiology and Immunology and the Walther Oncology Center, Indiana University School of Medicine and the Walther Cancer Institute, Indianapolis, IN 46202 USA.
| | | |
Collapse
|
41
|
Yang X, Lu P, Fujii C, Nakamoto Y, Gao JL, Kaneko S, Murphy PM, Mukaida N. Essential contribution of a chemokine, CCL3, and its receptor, CCR1, to hepatocellular carcinoma progression. Int J Cancer 2006; 118:1869-1876. [PMID: 16284949 DOI: 10.1002/ijc.21596] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We previously observed that a chemokine, macrophage inflammatory protein-1 alpha/CCL3, and its receptor, CCR1, were aberrantly expressed in human hepatocellular carcinoma (HCC) tissues. Here, we show that CCL3 and CCR1 are also expressed in 2 different models of this cancer; N-nitrosodiethylamine (DEN)-induced HCC and HCC induced by hepatitis B virus surface (HBs) antigen-primed splenocyte transfer to myelo-ablated syngeneic HBs antigen transgenic mice. At 10 months after DEN treatment, foci number and sizes were remarkably reduced in CCR1- and CCL3-deficient mice, compared with those of wild-type (WT) mice, although tumor incidence were marginally, but significantly, higher in CCR1- and CCL3-deficient mice than in WT mice. Of note is that tumor angiogenesis was also markedly diminished in CCL3- and CCR1-deficient mice, with a concomitant reduction in the number of intratumoral Kupffer cells, a rich source of growth factors and matrix metalloproteinases (MMPs). Among growth factors and MMPs that we examined, only MMP9 and MMP13 gene expression was augmented progressively in liver of WT mice after DEN treatment. Moreover, MMP9, but not MMP13, gene expression was attenuated in CCR1- and CCL3-deficient mice, compared with that of WT mice. Furthermore, MMP9 was expressed mainly by mononuclear cells but not hepatoma cells, and MMP9-expressing cell numbers were decreased in CCR1- or CCL3-deficient mice, compared with WT mice. These observations suggest the contribution of the CCR1-CCL3 axis to HCC progression.
Collapse
MESH Headings
- Alkylating Agents/administration & dosage
- Animals
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/physiopathology
- Chemokine CCL3
- Chemokine CCL4
- Diethylnitrosamine/administration & dosage
- Disease Models, Animal
- Disease Progression
- Gene Expression Profiling
- Hepatitis B virus
- Liver Neoplasms/genetics
- Liver Neoplasms/physiopathology
- Macrophage Inflammatory Proteins/biosynthesis
- Macrophage Inflammatory Proteins/genetics
- Male
- Matrix Metalloproteinases/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neovascularization, Pathologic
- Receptors, CCR1
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Spleen/cytology
Collapse
Affiliation(s)
- Xiaoqin Yang
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ishida N, Hayashi K, Hattori A, Yogo K, Kimura T, Takeya T. CCR1 acts downstream of NFAT2 in osteoclastogenesis and enhances cell migration. J Bone Miner Res 2006; 21:48-57. [PMID: 16355273 DOI: 10.1359/jbmr.051001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Revised: 10/04/2005] [Accepted: 10/05/2005] [Indexed: 11/18/2022]
Abstract
UNLABELLED We found that a chemokine receptor gene, CCR1, acts downstream of NFAT2 in RANKL-stimulated RAW264 and bone marrow cells. The upstream regulatory region of CCR1 showed RANKL-dependent and CsA-suppressible promoter activity. Downregulation of the expression and function of CCR1 suppressed cell migration. INTRODUCTION We previously reported that the expression of NFAT2 induced by RANKL is a key process for progression to multinucleated cells in an in vitro osteoclastogenesis system. Identifying the target genes of NFAT2 would thus be informative about the differentiation process. We focused here on chemokine and chemokine receptor genes that act downstream of NFAT2 in RAW264 cells as well as osteoclast precursors prepared from bone marrow cells. MATERIALS AND METHODS RAW264 mouse monocyte/macrophage line cells were cultured with or without cyclosporin A (CsA) in the presence of RANKL or glutathione S-transferase (GST). Osteoclast precursors were prepared from bone marrow cells. RANKL-inducible and CsA-suppressible genes were searched for by microarray analysis, and expression was confirmed by quantitative RT-PCR. Promoter activity was measured by luciferase gene reporter assay. Short interfering (si)RNA for CCR1 was introduced in RAW264 cells. Cell migration activity was examined using a Boyden chamber assay. RESULTS AND CONCLUSIONS We identified the chemokine receptor gene CCR1 as a gene showing significant differential expression profiles in osteoclastogenesis in the presence versus the absence of CsA, an inhibitor of NFAT. This property was unique to CCR1 among the chemokine and chemokine receptor genes examined in both RAW264 and bone marrow cells. The upstream regulatory region was isolated from CCR1, and its RANKL-dependent and CsA-suppressible promoter activity was confirmed. The functional significance of CCR1 was assessed by monitoring the migration of cells in a transwell migration assay, and this activity was abolished when either CsA- or CCR1 siRNA-treated cells were used. Moreover, treatment with a Galpha inhibitor pertussis toxin (PTX) or methiolynated-regulated on activation, normal T cells expressed and secreted (Met-RANTES), an antagonist of CCR1, suppressed multinucleated cell formation in the bone marrow cell system. Together, these results suggest that the CCR1 signaling cascade is under the control of NFAT2 and seems to enhance the migration of differentiating osteoclasts.
Collapse
Affiliation(s)
- Norihiro Ishida
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan
| | | | | | | | | | | |
Collapse
|
43
|
Shih CH, van Eeden SF, Goto Y, Hogg JC. CCL23/myeloid progenitor inhibitory factor-1 inhibits production and release of polymorphonuclear leukocytes and monocytes from the bone marrow. Exp Hematol 2005; 33:1101-8. [PMID: 16219532 DOI: 10.1016/j.exphem.2005.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 06/17/2005] [Accepted: 06/20/2005] [Indexed: 11/18/2022]
Abstract
OBJECTIVE CCL23/Myeloid progenitor inhibitory factor-1 is a human CC chemokine with potent in vitro suppressor effects on both human and murine myeloid progenitor cells. This study concerns in vivo inhibitory effect of CCL23 on production of polymorphonuclear leukocytes (PMNs) and monocytes in the bone marrow and their release into the circulation. METHODS 5'-Bromo-2'-deoxyuridine (BrdU; 100 mg/kg) was used to label dividing PMNs and monocytes in the bone marrow, and BrdU-labeled cells were followed for 10 days in the circulation and identified using immunocytochemistry. Rabbits were given CCL23 (100 mug/kg, n = 5) or saline (control: n = 5) intravenously daily for 3 days before labeling with BrdU. Turnover of PMNs and monocytes in the bone marrow and their transit times through the bone marrow were calculated. RESULTS CCL23 treatment tended to prolong transit time of PMN (98.4 +/- 4.3 hours vs 111.2 +/- 3.8 hours, control vs CCL23, p = 0.06) through the bone marrow and decreased the size of the bone marrow mitotic pool of PMN (p < 0.01). CCL23 treatment also prolonged the transit time of monocyte (43.4 +/- 3.1 hours vs 54.2 +/- 1.3 hours, control vs CCL23, p < 0.05) through the bone marrow and decreased turnover and pool size of monocytes in the bone marrow (p < 0.05). CONCLUSION We conclude that CCL23 suppresses PMN and monocyte progenitors, decreases the pool size and slows their turnover in the bone marrow.
Collapse
Affiliation(s)
- Chih-Horng Shih
- James Hogg iCAPTURE Centre for Cardiovascular and Pulmonary Research, University of British Columbia, St. Paul's Hospital, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
44
|
Yang M, Mailhot G, MacKay CA, Mason-Savas A, Aubin J, Odgren PR. Chemokine and chemokine receptor expression during colony stimulating factor-1-induced osteoclast differentiation in the toothless osteopetrotic rat: a key role for CCL9 (MIP-1gamma) in osteoclastogenesis in vivo and in vitro. Blood 2005; 107:2262-70. [PMID: 16304045 PMCID: PMC1895722 DOI: 10.1182/blood-2005-08-3365] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Osteoclasts differentiate from hematopoietic precursors under systemic and local controls. Chemokines and receptors direct leukocyte traffic throughout the body and may help regulate site-specific bone resorption. We investigated bone gene expression in vivo during rapid osteoclast differentiation induced by colony-stimulating factor 1 (CSF-1) in Csf1-null toothless (tl/tl) rats. Long-bone RNA from CSF-1-treated tl/tl rats was analyzed by high-density microarray over a time course. TRAP (tartrate-resistant acid phosphatase)-positive osteoclasts appeared on day 2, peaked on day 4, and decreased slightly on day 6, as marrow space was expanding. TRAP and cathepsin K mRNA paralleled the cell counts. We examined all chemokine and receptor mRNAs on the arrays. CCL9 was strongly induced and peaked on day 2, as did its receptor, CCR1, and regulatory receptors c-Fms (CSF-1 receptor) and RANK (receptor activator of nuclear factor kappaB). Other chemokines and receptors showed little or no significant changes. In situ hybridization and immunohistochemistry revealed CCL9 in small, immature osteoclasts on day 2 and in mature cells at later times. Anti-CCL9 antibody inhibited osteoclast differentiation in culture and significantly suppressed the osteoclast response in CSF-1-treated tl/tl rats. While various chemokines have been implicated in osteoclastogenesis in vitro, this first systematic analysis of chemokines and receptors during osteoclast differentiation in vivo highlights the key role of CCL9 in this process.
Collapse
Affiliation(s)
- Meiheng Yang
- Dept of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | |
Collapse
|
45
|
Gupta S, Schulz-Maronde S, Kutzleb C, Richter R, Forssmann WG, Kapp A, Forssmann U, Elsner J. Cloning, expression, and functional characterization of cynomolgus monkey (Macaca fascicularis) CC chemokine receptor 1. J Leukoc Biol 2005; 78:1175-84. [PMID: 16204626 DOI: 10.1189/jlb.0605326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The CC chemokine receptor 1 (CCR1) has emerged as a relevant factor contributing to inflammatory diseases such as allergic asthma. Commonly used animal models of allergic airway inflammation, especially murine models, have certain limitations. The elaborate, nonhuman, primate models of asthma display the highest comparability with the situation in humans. These models play an important role in the understanding of the pathogenesis of asthma. To improve the understanding in cynomolgus monkey models, we identified and characterized CCR1 in this nonhuman primate. Initially, we cloned the cynomolgus monkey CCR1 (cCCR1) gene, and the sequence analysis revealed high homology at the nucleotide (92%) and amino acid (88.4%) levels with its human counterpart. Human embryonic kidney 293 cells were stably transfected with cCCR1 and used in functional assays. Among those CCR1 ligands tested, CCL14(9-74) was most potent in the induction of intracellular Ca2+ fluxes as observed for human CCR1 (hCCR1). Complete cross-desensitization could be achieved between CCL14(9-74) and CCL15. However, CCL3 could not fully abrogate the response to the potent ligand CCL14(9-74). Competition-binding studies with radiolabeled CCL3 concordantly showed that CCL14(9-74) has a higher affinity to cCCR1 than hCCL3. Moreover, differential tissue-specific expression of cCCR1 was investigated by real-time quantitative polymerase chain reaction, displaying the highest levels in spleen. This study adds basic information needed for the evaluation of the role of CCR1 in the pathophysiology of asthma using the highly relevant cynomolgus monkey model and in addition, aids in the preclinical evaluation of potential novel drugs targeting CCR1.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Binding, Competitive
- Cell Line
- Chemokines/pharmacology
- Chemokines, CC/pharmacology
- Cloning, Molecular
- Disease Models, Animal
- GTP-Binding Protein alpha Subunits, Gi-Go/genetics
- Gene Expression Regulation
- Humans
- Ligands
- Macaca fascicularis/genetics
- Mice
- Molecular Sequence Data
- Rats
- Receptors, CCR1
- Receptors, Chemokine/drug effects
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Homology, Amino Acid
- Time Factors
Collapse
Affiliation(s)
- Shipra Gupta
- Department of Dermatology and Allergology, An-Institut, Hannover Medical School, Germany
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Chemokines (chemotactic cytokines) are a family of low-molecular-weight proteins that direct the cellular migration of leukocytes by binding to and activating the G protein-coupled receptors displayed on the leukocyte cell surface. The inadvertent or excessive generation of chemokines has been associated with the inflammatory component of several disease processes, and consequently, considerable efforts have been made to characterise chemokine/chemokine receptor interactions with the ultimate aim of therapeutic intervention. This review focuses on the biology of CC chemokine receptor 1, which together with its ligands is thought to recruit leukocytes during the progression of rheumatoid arthritis, multiple sclerosis and organ transplant rejection. The developments made in antagonising this receptor and efficacies of these compounds in the clinical setting are also highlighted.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Drug Evaluation, Preclinical/trends
- Drugs, Investigational/chemistry
- Drugs, Investigational/pharmacology
- Drugs, Investigational/therapeutic use
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Humans
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/metabolism
- Receptors, CCR1
- Receptors, Chemokine/antagonists & inhibitors
- Receptors, Chemokine/metabolism
- Technology, Pharmaceutical/trends
Collapse
Affiliation(s)
- James E Pease
- Biomedical Sciences Division, Faculty of Medicine, Imperial College of Science, Technology and Medicine, South Kensington Campus, London SW7 2AZ, UK.
| | | |
Collapse
|
47
|
Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper S, Plett PA, Liles WC, Li X, Graham-Evans B, Campbell TB, Calandra G, Bridger G, Dale DC, Srour EF. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. ACTA ACUST UNITED AC 2005; 201:1307-18. [PMID: 15837815 PMCID: PMC2213145 DOI: 10.1084/jem.20041385] [Citation(s) in RCA: 868] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Improving approaches for hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC) mobilization is clinically important because increased numbers of these cells are needed for enhanced transplantation. Chemokine stromal cell derived factor-1 (also known as CXCL12) is believed to be involved in retention of HSCs and HPCs in bone marrow. AMD3100, a selective antagonist of CXCL12 that binds to its receptor, CXCR4, was evaluated in murine and human systems for mobilizing capacity, alone and in combination with granulocyte colony-stimulating factor (G-CSF). AMD3100 induced rapid mobilization of mouse and human HPCs and synergistically augmented G-CSF-induced mobilization of HPCs. AMD3100 also mobilized murine long-term repopulating (LTR) cells that engrafted primary and secondary lethally-irradiated mice, and human CD34(+) cells that can repopulate nonobese diabetic-severe combined immunodeficiency (SCID) mice. AMD3100 synergized with G-CSF to mobilize murine LTR cells and human SCID repopulating cells (SRCs). Human CD34(+) cells isolated after treatment with G-CSF plus AMD3100 expressed a phenotype that was characteristic of highly engrafting mouse HSCs. Synergy of AMD3100 and G-CSF in mobilization was due to enhanced numbers and perhaps other characteristics of the mobilized cells. These results support the hypothesis that the CXCL12-CXCR4 axis is involved in marrow retention of HSCs and HPCs, and demonstrate the clinical potential of AMD3100 for HSC mobilization.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
A proinflammatory chemokine, CCL3, sensitizes the heat- and capsaicin-gated ion channel TRPV1. Proc Natl Acad Sci U S A 2005. [PMID: 15764707 DOI: 10.1073/pnas/0406030102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pain, a critical component of host defense, is one hallmark of the inflammatory response. We therefore hypothesized that pain might be exacerbated by proinflammatory chemokines. To test this hypothesis, CCR1 was cotransfected into human embryonic kidney (HEK)293 cells together with transient receptor potential vanilloid 1 (TRPV1), a cation channel required for certain types of thermal hyperalgesia. In these cells, capsaicin and anandamide induced Ca(2+) influx mediated by TRPV1. When CCR1:TRPV1/HEK293 cells were pretreated with CCL3, the sensitivity of TRPV1, as indicated by the Ca(2+) influx, was increased approximately 3-fold. RT-PCR analysis showed that a spectrum of chemokine and cytokine receptors is expressed in rat dorsal root ganglia (DRG). Immunohistochemical staining of DRG showed that CCR1 is coexpressed with TRPV1 in >85% of small-diameter neurons. CCR1 on DRG neurons was functional, as demonstrated by CCL3-induced Ca(2+) ion influx and PKC activation. Pretreatment with CCL3 enhanced the response of DRG neurons to capsaicin or anandamide. This sensitization was inhibited by pertussis toxin, U73122, or chelerythrine chloride, inhibitors of Gi-protein, phospholipase C, and protein kinase C, respectively. Intraplantar injection of mice with CCL3 decreased their hot-plate response latency. That a proinflammatory chemokine, by interacting with its receptor on small-diameter neurons, sensitizes TRPV1 reveals a previously undescribed mechanism of receptor cross-sensitization that may contribute to hyperalgesia during inflammation.
Collapse
|
49
|
Zhang N, Inan S, Inan S, Cowan A, Sun R, Wang JM, Rogers TJ, Caterina M, Oppenheim JJ. A proinflammatory chemokine, CCL3, sensitizes the heat- and capsaicin-gated ion channel TRPV1. Proc Natl Acad Sci U S A 2005; 102:4536-41. [PMID: 15764707 PMCID: PMC555471 DOI: 10.1073/pnas.0406030102] [Citation(s) in RCA: 192] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Pain, a critical component of host defense, is one hallmark of the inflammatory response. We therefore hypothesized that pain might be exacerbated by proinflammatory chemokines. To test this hypothesis, CCR1 was cotransfected into human embryonic kidney (HEK)293 cells together with transient receptor potential vanilloid 1 (TRPV1), a cation channel required for certain types of thermal hyperalgesia. In these cells, capsaicin and anandamide induced Ca(2+) influx mediated by TRPV1. When CCR1:TRPV1/HEK293 cells were pretreated with CCL3, the sensitivity of TRPV1, as indicated by the Ca(2+) influx, was increased approximately 3-fold. RT-PCR analysis showed that a spectrum of chemokine and cytokine receptors is expressed in rat dorsal root ganglia (DRG). Immunohistochemical staining of DRG showed that CCR1 is coexpressed with TRPV1 in >85% of small-diameter neurons. CCR1 on DRG neurons was functional, as demonstrated by CCL3-induced Ca(2+) ion influx and PKC activation. Pretreatment with CCL3 enhanced the response of DRG neurons to capsaicin or anandamide. This sensitization was inhibited by pertussis toxin, U73122, or chelerythrine chloride, inhibitors of Gi-protein, phospholipase C, and protein kinase C, respectively. Intraplantar injection of mice with CCL3 decreased their hot-plate response latency. That a proinflammatory chemokine, by interacting with its receptor on small-diameter neurons, sensitizes TRPV1 reveals a previously undescribed mechanism of receptor cross-sensitization that may contribute to hyperalgesia during inflammation.
Collapse
Affiliation(s)
- Ning Zhang
- Laboratory of Molecular Immunoregulation, Intramural Research Support Program, Division of Basic Sciences, National Cancer Institute, National Institutes of Health, Frederick, MD 21702-1201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
de Mendonça FL, da Fonseca PCA, Phillips RM, Saldanha JW, Williams TJ, Pease JE. Site-directed mutagenesis of CC chemokine receptor 1 reveals the mechanism of action of UCB 35625, a small molecule chemokine receptor antagonist. J Biol Chem 2004; 280:4808-16. [PMID: 15548526 DOI: 10.1074/jbc.m412267200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The chemokine receptor CCR1 and its principal ligand, CCL3/MIP-1alpha, have been implicated in the pathology of several inflammatory diseases including rheumatoid arthritis, multiple sclerosis, and asthma. As such, these molecules are the focus of much research with the ultimate aim of developing novel therapies. We have described previously a non-competitive small molecule antagonist of CCR1 (UCB 35625), which we hypothesized interacted with amino acids located within the receptor transmembrane (TM) helices (Sabroe, I., Peck, M. J., Jan Van Keulen, B., Jorritsma, A., Simmons, G., Clapham, P. R., Williams, T. J., and Pease, J. E. (2000) J. Biol. Chem. 275, 25985-25992). Here we describe an approach to identifying the mechanism by which the molecule antagonizes CCR1. Thirty-three point mutants of CCR1 were expressed transiently in L1.2 cells, and the cells were assessed for their capacity to migrate in response to CCL3 in the presence or absence of UCB 35625. Cells expressing the mutant constructs Y41A (TM helix 1, or TM1), Y113A (TM3), and E287A (TM7) were responsive to CCL3 but resistant to the antagonist, consistent with a role for the TM helices in CCR1 interactions with UCB 35625. Subsequent molecular modeling successfully docked the compound with CCR1 and suggests that the antagonist ligates TM1, 2, and 7 of CCR1 and severely impedes access to TM2 and TM3, a region thought to be perturbed by the chemokine amino terminus during the process of receptor activation. Insights into the mechanism of action of these compounds may facilitate the development of more potent antagonists that show promise as future therapeutic agents in the treatment of inflammatory disease.
Collapse
MESH Headings
- Amino Acid Sequence
- Binding Sites
- Binding, Competitive
- Cell Membrane/metabolism
- Cell Movement
- Chemotaxis
- Dose-Response Relationship, Drug
- Glutamic Acid/chemistry
- Humans
- Inflammation
- Models, Chemical
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Point Mutation
- Protein Binding
- Protein Conformation
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Receptors, CCR1
- Receptors, Chemokine/chemistry
- Receptors, Chemokine/genetics
- Sequence Homology, Amino Acid
- Signal Transduction
- Software
- Stereoisomerism
- Transfection
- Xanthenes/pharmacology
Collapse
Affiliation(s)
- Filipa Lopes de Mendonça
- Leukocyte Biology, Faculty of Medicine, Sir Alexander Fleming Building, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | | | | | | | | | | |
Collapse
|