1
|
Capitano ML, Sammour Y, Ropa J, Legendre M, Mor‐Vaknin N, Markovitz DM. DEK, a nuclear protein, is chemotactic for hematopoietic stem/progenitor cells acting through CXCR2 and Gαi signaling. J Leukoc Biol 2022; 112:449-456. [PMID: 35137444 PMCID: PMC9541944 DOI: 10.1002/jlb.3ab1120-740r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022] Open
Abstract
Few cytokines/growth modulating proteins are known to be chemoattractants for hematopoietic stem (HSC) and progenitor cells (HPC); stromal cell-derived factor 1α (SDF1α/CXCL12) being the most potent known such protein. DEK, a nuclear DNA-binding chromatin protein with hematopoietic cytokine-like activity, is a chemotactic factor attracting mature immune cells. Transwell migration assays were performed to test whether DEK serves as a chemotactic agent for HSC/HPC. DEK induced dose- and time-dependent directed migration of lineage negative (Lin- ) Sca-1+ c-Kit+ (LSK) bone marrow (BM) cells, HSCs and HPCs. Checkerboard assays demonstrated that DEK's activity was chemotactic (directed), not chemokinetic (random migration), in nature. DEK and SDF1α compete for HSC/HPC chemotaxis. Blocking CXCR2 with neutralizing antibodies or inhibiting Gαi protein signaling with Pertussis toxin pretreatment inhibited migration of LSK cells toward DEK. Thus, DEK is a novel and rare chemotactic agent for HSC/HPC acting in a direct or indirect CXCR2 and Gαi protein-coupled signaling-dependent manner.
Collapse
Affiliation(s)
- Maegan L. Capitano
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yasser Sammour
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - James Ropa
- Department of Microbiology and ImmunologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Maureen Legendre
- Department of Internal Medicine, Division of Infectious DiseasesUniversity of MichiganAnn ArborMichiganUSA
| | - Nirit Mor‐Vaknin
- Department of Internal Medicine, Division of Infectious DiseasesUniversity of MichiganAnn ArborMichiganUSA
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious DiseasesUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
2
|
Capitano ML, Mor-Vaknin N, Saha AK, Cooper S, Legendre M, Guo H, Contreras-Galindo R, Kappes F, Sartor MA, Lee CT, Huang X, Markovitz DM, Broxmeyer HE. Secreted nuclear protein DEK regulates hematopoiesis through CXCR2 signaling. J Clin Invest 2019; 129:2555-2570. [PMID: 31107242 PMCID: PMC6546479 DOI: 10.1172/jci127460] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
The nuclear protein DEK is an endogenous DNA-binding chromatin factor regulating hematopoiesis. DEK is one of only 2 known secreted nuclear chromatin factors, but whether and how extracellular DEK regulates hematopoiesis is not known. We demonstrated that extracellular DEK greatly enhanced ex vivo expansion of cytokine-stimulated human and mouse hematopoietic stem cells (HSCs) and regulated HSC and hematopoietic progenitor cell (HPC) numbers in vivo and in vitro as determined both phenotypically (by flow cytometry) and functionally (through transplantation and colony formation assays). Recombinant DEK increased long-term HSC numbers and decreased HPC numbers through a mechanism mediated by the CXC chemokine receptor CXCR2 and heparan sulfate proteoglycans (HSPGs) (as determined utilizing Cxcr2-/- mice, blocking CXCR2 antibodies, and 3 different HSPG inhibitors) that was associated with enhanced phosphorylation of ERK1/2, AKT, and p38 MAPK. To determine whether extracellular DEK required nuclear function to regulate hematopoiesis, we utilized 2 mutant forms of DEK: one that lacked its nuclear translocation signal and one that lacked DNA-binding ability. Both altered HSC and HPC numbers in vivo or in vitro, suggesting the nuclear function of DEK is not required. Thus, DEK acts as a hematopoietic cytokine, with the potential for clinical applicability.
Collapse
Affiliation(s)
- Maegan L. Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Anjan K. Saha
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Maureen Legendre
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Haihong Guo
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Rafael Contreras-Galindo
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Ferdinand Kappes
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher T. Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Hal E. Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
3
|
Chen X, Li B, Gao Y, Ji J, Wu Z, Chen S. Saponins from Sanguisorba officinalis Improve Hematopoiesis by Promoting Survival through FAK and Erk1/2 Activation and Modulating Cytokine Production in Bone Marrow. Front Pharmacol 2017; 8:130. [PMID: 28360858 PMCID: PMC5353277 DOI: 10.3389/fphar.2017.00130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/01/2017] [Indexed: 12/31/2022] Open
Abstract
Radix Sanguisorbae, the root of Sanguisorba officinalis L. is used as traditional Chinese medicine. In recent decades, it has been reported to be clinically effective against myelosuppression induced by chemotherapy and/ or radiotherapy. However, the underlining mechanism has not been well studied. In this work, we evaluated the hematopoietic effect of total saponins from S. officinalis L. on myelosuppressive mice induced by cyclophosphamide and by60Co-γ-irradiation and confirmed the therapeutic effect. Then, we found total saponins and their characteristic constituents Ziyuglycoside I and Ziyuglycoside II can inhibit apoptosis of TF-1 cells caused by cytokine deprivation, and promote survival of mouse bone marrow nuclear cells through focal adhesion kinase (FAK) and extracellular signal-regulated kinase 1/2 (Erk1/2) activation in vitro. In addition, they can down-regulate macrophage inflammatory protein 2 (MIP-2), platelet factor 4 (PF4) and P-selectin secretion, which are reported to be suppressive to hematopoiesis, both in vitro and in vivo. These results suggest that promotion of survival through FAK and Erk1/2 activation and inhibition of suppressive cytokines in the bone marrow is likely to be the pharmacological mechanism underlying the hematopoietic effect of saponins from S. officinalis L.
Collapse
Affiliation(s)
- Xin Chen
- Chengdu Institute of Biology, Chinese Academy of Sciences (CAS)Sichuan, China; Graduate School, University of Chinese Academy of Sciences (CAS)Beijing, China
| | - Bogang Li
- Chengdu Institute of Biology, Chinese Academy of Sciences (CAS)Sichuan, China; Di Ao Pharmaceutical GroupSichuan, China
| | - Yue Gao
- Institute of Radiation Medicine, Academy of Military Medical Sciences Beijing, China
| | - Jianxin Ji
- Chengdu Institute of Biology, Chinese Academy of Sciences (CAS) Sichuan, China
| | - Zhongliu Wu
- Chengdu Institute of Biology, Chinese Academy of Sciences (CAS) Sichuan, China
| | - Shuang Chen
- Chengdu Institute of Biology, Chinese Academy of Sciences (CAS) Sichuan, China
| |
Collapse
|
4
|
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
|
5
|
Nakayama R, Kuroda J, Taniyama N, Yamamoto-Sugitani M, Wada S, Kiyota M, Mizutani S, Chinen Y, Matsumoto Y, Nagoshi H, Shimura Y, Kobayashi T, Horiike S, Sato K, Taniwaki M. Suppression of SERPINA1-albumin complex formation by galectin-3 overexpression leads to paracrine growth promotion of chronic myelogenous leukemia cells. Leuk Res 2013; 38:103-8. [PMID: 23953881 DOI: 10.1016/j.leukres.2013.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/12/2013] [Indexed: 01/08/2023]
Abstract
Galectin-3 is induced in chronic myelogenous leukemia (CML) cells by co-culture with bone marrow stromal cells, making paracrine growth promotion of CML cells in conditioned medium (CM) from galectin-3 overexpressing CML cells more potent. We used gel filtration chromatography to demonstrate that the bovine SERPINA1-fetal bovine serum albumin (BSA) complex was specifically suppressed in CM from galectin-3 overexpressing cells. The SERPINA1-BSA complex as well as human plasma SERPINA1 inhibited the growth of CML cells, while exogenous galectin-3 partly offset this effect. These findings suggest that galectin-3 overexpression promotes paracrine growth of CML cells by interfering with the action of the growth inhibitory SERPINA1-albumin complex.
Collapse
Affiliation(s)
- Ryuko Nakayama
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Nobuko Taniyama
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Mio Yamamoto-Sugitani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sayori Wada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Miki Kiyota
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shinsuke Mizutani
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshiaki Chinen
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Matsumoto
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisao Nagoshi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Shimura
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsutomu Kobayashi
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeo Horiike
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Sato
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Masafumi Taniwaki
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
6
|
Rapid expansion of human hematopoietic stem cells by automated control of inhibitory feedback signaling. Cell Stem Cell 2012; 10:218-29. [PMID: 22305571 DOI: 10.1016/j.stem.2012.01.003] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 12/15/2011] [Accepted: 01/06/2012] [Indexed: 12/28/2022]
Abstract
Clinical hematopoietic transplantation outcomes are strongly correlated with the numbers of cells infused. Anticipated novel therapeutic implementations of hematopoietic stem cells (HSCs) and their derivatives further increase interest in strategies to expand HSCs ex vivo. A fundamental limitation in all HSC-driven culture systems is the rapid generation of differentiating cells and their secreted inhibitory feedback signals. Herein we describe an integrated computational and experimental strategy that enables a tunable reduction in the global levels and impact of paracrine signaling factors in an automated closed-system process by employing a controlled fed-batch media dilution approach. Application of this system to human cord blood cells yielded a rapid (12-day) 11-fold increase of HSCs with self-renewing, multilineage repopulating ability. These results highlight the marked improvements that control of feedback signaling can offer primary stem cell culture and demonstrate a clinically relevant rapid and relatively low culture volume strategy for ex vivo HSC expansion.
Collapse
|
7
|
Moldenhauer A, Futschik M, Lu H, Helmig M, Götze P, Bal G, Zenke M, Han W, Salama A. Interleukin 32 promotes hematopoietic progenitor expansion and attenuates bone marrow cytotoxicity. Eur J Immunol 2011; 41:1774-86. [DOI: 10.1002/eji.201040986] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 01/23/2011] [Accepted: 03/15/2011] [Indexed: 11/07/2022]
|
8
|
Newman MB, Bakay RAE. Therapeutic potentials of human embryonic stem cells in Parkinson's disease. Neurotherapeutics 2008; 5:237-51. [PMID: 18394566 PMCID: PMC5084166 DOI: 10.1016/j.nurt.2008.02.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The loss of dopaminergic neurons of the substantia nigra is the pathological hallmark characteristic of Parkinson's disease (PD). The strategy of replacing these degenerating neurons with other cells that produce dopamine has been the main approach in the cell transplantation field for PD research. The isolation, differentiation, and long-term cultivation of human embryonic stem cells and the therapeutic research discovery made in relation to the beneficial properties of neurotrophic and neural growth factors has advanced the transplantation field beyond dopamine-producing cells. The present review addresses recent advances in human embryonic stem cell experimentation in relation to treating PD, as well as cell transplantation techniques in conjunction with alternative therapeutics.
Collapse
Affiliation(s)
- Mary B Newman
- Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
9
|
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
|
10
|
Broxmeyer HE, Cooper S, Hangoc G, Chang CH. Class II transactivator-mediated regulation of major histocompatibility complex class II antigen expression is important for hematopoietic progenitor cell suppression by chemokines and iron-binding proteins. Exp Hematol 2006; 34:1078-84. [PMID: 16863914 DOI: 10.1016/j.exphem.2006.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Iron-binding proteins H-ferritin (HF) and lactoferrin (LF), as well as chemokines, tumor necrosis factor (TNF)-alpha, and interferon (IFN)-gamma suppress hematopoietic progenitor cell (HPC) proliferation. Major histocompatibility complex (MHC) class II antigens have been associated with suppressive effects of HF and LF. Because the transcription factor class II transactivator (CIITA) regulates expression of MHC class II antigens, we evaluated influences of CIITA and MHC class II antigens on suppression of colony formation by murine bone marrow HPC in response to HF, LF, CC, and CXC chemokines, TNF-alpha, and IFN-gamma. We also evaluated hematopoiesis in mice deficient in both CIITA and MHC class II antigens (CIITA -/-), in mice deficient in MHC class II antigens but not in CIITA (MHC class II -/-), and in mice deficient in CIITA but not in MHC class II antigens (CIITA-IE). MATERIALS AND METHODS HF, LF, CCL3/MIP-1alpha, CXCL5/ENA-78, CXCL8/IL-8, CCL5/RANTES, TNF-alpha, and IFN-gamma were assessed for effects on colony formation by bone marrow HPC (colony-forming unit granulocyte-macrophage, burst-forming unit erythroid, and colony-forming unit multipotential) stimulated in vitro by combinations of growth factors including erythropoietin, stem cell factor, pokeweed mitogen mouse spleen cell conditioned medium, and hemin. Bone marrow cells were from CIITA -/-, MHC class II antigen -/-, CIITA-IE, and littermate control mice. We also evaluated cycling status (percent cells in S-phase) and absolute numbers of marrow and spleen HPC in these mice. RESULTS Multiple growth factor-stimulated colony formation by control bone marrow HPC was significantly suppressed by HF, LF, CCL3, CXCL5, CXCL8, TNF-alpha, and IFN-gamma, but not by CCL5. However, HPC from CIITA -/- and MHC class II antigen -/- mouse marrow was insensitive to inhibition by HF, LF, CCL3, CXCL5, CXCL8, and CCL5; these HPC were inhibited by TNF-alpha and IFN-gamma. Restoration of MHC class II expression in CIITA -/- (CIITA-IE) mice restored responsiveness of HPC to inhibition by HF, LF, CCL3, CXCL5, and CXCL8. Increased cycling of splenic HPC in CIITA -/- and MHC class II antigen -/-, compared to control and CIITA-IE, mice was noted. CONCLUSIONS Myelosuppressive effects of iron-binding proteins HF and LF and chemokines CCL3, CXCL5, and CXCL8 on mouse bone marrow HPC require expression of MHC class II antigens, and CIITA is involved in this responsiveness through its regulation of expression of MHC class II antigens.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Departments of Microbiology and Immunology, and the Walther Oncology Center, Indiana University School of Medicine, and the Walther Cancer Institute, Indianapolis, IN 46202-5181, USA.
| | | | | | | |
Collapse
|
11
|
Kwon BS, Hurtado JC, Lee ZH, Kwack KB, Seo SK, Choi BK, Koller BH, Wolisi G, Broxmeyer HE, Vinay DS. Immune responses in 4-1BB (CD137)-deficient mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:5483-90. [PMID: 12023342 DOI: 10.4049/jimmunol.168.11.5483] [Citation(s) in RCA: 169] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The 4-1BB (a TNFR superfamily member) is an inducible costimulatory molecule that can exert regulatory effects on T cells independently of CD28 stimulation. The in vitro expression of 4-1BB (CD137) is induced following activation of T cells with various stimuli, including anti-TCR mAbs, lectins, and a combination of PMA and ionomycin. To delineate further the physiological role of 4-1BB in immunity, mice deficient in this receptor were generated. These mutant mice developed normally, and were viable and fertile. Humoral responses to vesicular stomatitis virus were comparable with those seen in wild-type mice, whereas the IgG2a and IgG3 isotype responses to keyhole limpet hemocyanin were somewhat reduced in the mutant mice. The 4-1BB-deficient mice demonstrated enhanced T cell proliferation in response to mitogens or anti-CD3 even in the environment of reduced ability to secrete growth-supporting cytokines (IL-2 and IL-4). Although T cells from 4-1BB-deficient mice showed enhanced proliferation, the T cell immune responses of these animals, such as cytokine production and CTL activity, were diminished. In addition, 4-1BB deletion appears to play a role in the regulation of myeloid progenitor cell growth, leading to an increase in these precursor cells in peripheral blood, bone marrow, and spleen.
Collapse
Affiliation(s)
- Byoung S Kwon
- Immunomodulation Research Center, University of Ulsan, Ulsan, Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Chemokines, originally designated as chemoattractant cytokines, comprise a large family of molecules that have been implicated in a number of different functions mediated through chemokine receptors. Among these functions are regulatory roles in hematopoiesis that encompass effects on the proliferation, survival, and homing/migration of myeloid progenitor cells. This article reviews the field of chemokine regulation of hematopoiesis at the level of myeloid progenitor cells.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine and the Walther Cancer Institute, Indianapolis 46202-5254, USA.
| |
Collapse
|
13
|
Guest I, Uetrecht J. Bone marrow stem cell protection from chemotherapy by low--molecular-weight compounds. Exp Hematol 2001; 29:123-37. [PMID: 11166451 DOI: 10.1016/s0301-472x(00)00621-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The stem cells of the bone marrow have the capacity for both self-renewal and derivation of all the blood cell lineages. Consequently, toxicity to these cells can result in neutropenia, agranulocytosis, thrombocytopenia, pancytopenia, or aplastic anemia. Many anticancer drugs adversely affect the bone marrow, and neutropenia is a common limiting factor in dose escalation. In this review, we discuss agents that appear to have potential as bone marrow sparing agents. Computerized catalogs of the National Library of Medicine and Medline were searched for reports on low-molecular-weight compounds that detailed effects on the hematopoietic progenitor cells. The most promising agents are the endogenous peptides p-glutamic acid-glutamic acid-aspartic acid-cysteine-lysine and acetyl-serine-aspartic acid-lysine-proline, and the exogenous compounds amifostine and ammonium trichloro[dioxoethylene-O,O']tellurate, but several others are also discussed. These compounds preserve stem cell function in the presence of antineoplastic drugs of diverse pharmacological classes, and they do so by various mechanisms of action. Their present status in clinical practice is also detailed. More needs to be learned about their mechanisms of action and therapeutic potential, but the results are encouraging for some of these compounds and more clinical trials should be expected.
Collapse
Affiliation(s)
- I Guest
- Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
14
|
Abstract
Chemokines are a family of related proteins that regulate leukocyte infiltration into inflamed tissue and play important roles in disease processes. Among the biologic activities of chemokines is inhibition of proliferation of normal hematopoietic progenitors. However, chemokines that inhibit normal progenitors rarely inhibit proliferation of hematopoietic progenitors from patients with chronic myelogenous leukemia (CML). We and others recently cloned a subfamily of CC chemokines that share similar amino-terminal peptide sequences and a remarkable ability to chemoattract T cells. These chemokines, Exodus-1/LARC/MIP-3, Exodus-2/SLC/6Ckine/TCA4, and Exodus-3/CKβ11/MIP-3β, were found to inhibit proliferation of normal human marrow progenitors. The study described here found that these chemokines also inhibited the proliferation of progenitors in every sample of marrow from patients with CML that was tested. This demonstration of consistent inhibition of CML progenitor proliferation makes the 3 Exodus chemokines unique among chemokines.
Collapse
|
15
|
Grafte-Faure S, Leveque C, Ketata E, Jean P, Vasse M, Soria C, Vannier JP. Recruitment of primitive peripheral blood cells: synergism of interleukin 12 with interleukin 6 and stromal cell-derived FACTOR-1. Cytokine 2000; 12:1-7. [PMID: 10623435 DOI: 10.1006/cyto.1999.0520] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In bone marrow, haematopoietic stem cells (HSC) rely on close contact with stromal cells for proliferation and differentiation. Stromal cell-derived factor (SDF-1) is a chemokine produced by bone marrow stromal cells and has been reported to be a chemoattractant for CD34(+)cells. SDF-1 was evaluated for effects on proliferation of both mature and immature human progenitor cells in vitro. Neither proliferation nor maturation of peripheral blood cells was stimulated by SDF-1 alone. Moreover, we have previously demonstrated that 5-fluorouracile (5-FU) resistant HSC require a combination of interleukin 12 (IL-12), IL-6 and SCF for the production of morphologically recognizable clonogenic elements at day 14 in semisolid medium. Our data reported a strong enhancement of the IL-6, IL-12, SCF-induced synergism (172%) by SDF-1 (296.5%). Furthermore, our data suggest that this chemokine alone had no effect on triggering quiescent cells and may preserve these cells from 5-FU cell damage or upregulate early-acting cytokine receptors. Thus, SDF-1 might play a key role in early human haematopoiesis through its potent synergistic effects in combination with early-acting cytokines. These results suggest that a programmed response to sequential cytokine stimulation may be part of a control mechanism required for maintenance of proliferation of primitive HSC.
Collapse
Affiliation(s)
- S Grafte-Faure
- Research Department of Microenvironment and Cell Integrated Renewal (MERCI), 22, Boulevard Gambetta, Rouen Cedex, 76183, France.
| | | | | | | | | | | | | |
Collapse
|
16
|
Segerer S, Nelson PJ, Schlöndorff D. Chemokines, chemokine receptors, and renal disease: from basic science to pathophysiologic and therapeutic studies. J Am Soc Nephrol 2000; 11:152-176. [PMID: 10616852 DOI: 10.1681/asn.v111152] [Citation(s) in RCA: 346] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Leukocyte trafficking from peripheral blood into affected tissues is an essential component of the inflammatory reaction to virtually all forms of injury and is an important factor in the development of many kidney diseases. Advances in the past few years have highlighted the central role of a family of chemotactic cytokines called chemokines in this process. Chemokines help to control the selective migration and activation of inflammatory cells into injured renal tissue. Chemokines and their receptors are expressed by intrinsic renal cells as well as by infiltrating cells during renal inflammation. This study summarizes the in vitro and in vivo data on chemokines and chemokine receptors in renal diseases with a special focus on potential therapeutic effects on inflammatory processes.
Collapse
Affiliation(s)
- Stephan Segerer
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| | - Peter J Nelson
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| | - Detlef Schlöndorff
- Medizinische Poliklinik, Klinikum Innenstadt der Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
17
|
Differentiation Stage–Specific Regulation of Primitive Human Hematopoietic Progenitor Cycling by Exogenous and Endogenous Inhibitors in an In Vivo Model. Blood 1999. [DOI: 10.1182/blood.v94.11.3722] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice transplanted with human cord blood or adult marrow cells and injected 6 weeks posttransplant with 2 daily doses of transforming growth factor-β1 (TGF-β1), monocyte chemoattractant protein-1 (MCP-1), or a nonaggregating form of macrophage inflammatory protein-1 (MIP-1) showed unique patterns of inhibition of human progenitor proliferation 1 day later. TGF-β1 was active on long-term culture initiating cells (LTC-IC) and on primitive erythroid and granulopoietic colony-forming cells (HPP-CFC), but had no effect on mature CFC. MCP-1 inhibited the cycling of both types of HPP-CFC but not LTC-IC. MIP-1 did not inhibit either LTC-IC or granulopoietic HPP-CFC but was active on erythroid HPP-CFC and mature granulopoietic CFC. All of these responses were independent of the source of human cells transplanted. LTC-IC of either human cord blood or adult marrow origin continue to proliferate in NOD/SCID mice for many weeks, although the turnover of all types of human CFC in mice transplanted with adult human marrow (but not cord blood) is downregulated after 6 weeks. Interestingly, administration of either MIP-1β, an antagonist of both MIP-1 and MCP-1 or MCP-1(9-76), an antagonist of MCP-1 (and MCP-2 and MCP-3), into mice in which human marrow-derived CFC had become quiescent, caused the rapid reactivation of these progenitors in vivo. These results provide the first definition of stage-specific inhibitors of human hematopoietic progenitor cell cycling in vivo. In addition they show that endogenous chemokines can contribute to late graft failure, which can be reversed by the administration of specific antagonists.
Collapse
|
18
|
Kim CH, Hangoc G, Cooper S, Helgason CD, Yew S, Humphries RK, Krystal G, Broxmeyer HE. Altered responsiveness to chemokines due to targeted disruption of SHIP. J Clin Invest 1999; 104:1751-9. [PMID: 10606629 PMCID: PMC409879 DOI: 10.1172/jci7310] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/1999] [Accepted: 10/26/1999] [Indexed: 11/17/2022] Open
Abstract
SHIP has been implicated in negative signaling in a number of hematopoietic cell types and is postulated to downregulate phosphatidylinositol-3-kinase- (PI-3K-) initiated events in diverse receptor signaling pathways. Because PI-3K is implicated in chemokine signaling, we investigated whether SHIP plays any role in cellular responses to chemokines. We found that a number of immature and mature hematopoietic cells from SHIP-deficient mice manifested enhanced directional migration (chemotaxis) in response to the chemokines stromal cell-derived factor-1 (SDF-1) and B-lymphocyte chemoattractant (BLC). SHIP(-/-) cells were also more active in calcium influx and actin polymerization in response to SDF-1. However, colony formation by SHIP-deficient hematopoietic progenitor cell (HPCs) was not inhibited by 13 myelosuppressive chemokines that normally inhibit proliferation of HPCs. These altered biologic activities of chemokines on SHIP-deficient cells are not caused by simple modulation of chemokine receptor expression in SHIP-deficient mice, implicating SHIP in the modulation of chemokine-induced signaling and downstream effects.
Collapse
Affiliation(s)
- C H Kim
- Departments of Microbiology/Immunology and Medicine and the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Differentiation Stage–Specific Regulation of Primitive Human Hematopoietic Progenitor Cycling by Exogenous and Endogenous Inhibitors in an In Vivo Model. Blood 1999. [DOI: 10.1182/blood.v94.11.3722.423k20_3722_3729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice transplanted with human cord blood or adult marrow cells and injected 6 weeks posttransplant with 2 daily doses of transforming growth factor-β1 (TGF-β1), monocyte chemoattractant protein-1 (MCP-1), or a nonaggregating form of macrophage inflammatory protein-1 (MIP-1) showed unique patterns of inhibition of human progenitor proliferation 1 day later. TGF-β1 was active on long-term culture initiating cells (LTC-IC) and on primitive erythroid and granulopoietic colony-forming cells (HPP-CFC), but had no effect on mature CFC. MCP-1 inhibited the cycling of both types of HPP-CFC but not LTC-IC. MIP-1 did not inhibit either LTC-IC or granulopoietic HPP-CFC but was active on erythroid HPP-CFC and mature granulopoietic CFC. All of these responses were independent of the source of human cells transplanted. LTC-IC of either human cord blood or adult marrow origin continue to proliferate in NOD/SCID mice for many weeks, although the turnover of all types of human CFC in mice transplanted with adult human marrow (but not cord blood) is downregulated after 6 weeks. Interestingly, administration of either MIP-1β, an antagonist of both MIP-1 and MCP-1 or MCP-1(9-76), an antagonist of MCP-1 (and MCP-2 and MCP-3), into mice in which human marrow-derived CFC had become quiescent, caused the rapid reactivation of these progenitors in vivo. These results provide the first definition of stage-specific inhibitors of human hematopoietic progenitor cell cycling in vivo. In addition they show that endogenous chemokines can contribute to late graft failure, which can be reversed by the administration of specific antagonists.
Collapse
|
20
|
Kim CH, Qu CK, Hangoc G, Cooper S, Anzai N, Feng GS, Broxmeyer HE. Abnormal chemokine-induced responses of immature and mature hematopoietic cells from motheaten mice implicate the protein tyrosine phosphatase SHP-1 in chemokine responses. J Exp Med 1999; 190:681-90. [PMID: 10477552 PMCID: PMC2195618 DOI: 10.1084/jem.190.5.681] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/1999] [Accepted: 07/01/1999] [Indexed: 11/04/2022] Open
Abstract
Chemokines regulate a number of biological processes, including trafficking of diverse leukocytes and proliferation of myeloid progenitor cells. SHP-1 (Src homology 2 domain tyrosine phosphatase 1), a phosphotyrosine phosphatase, is considered an important regulator of signaling for a number of cytokine receptors. Since specific tyrosine phosphorylation of proteins is important for biological activities induced by chemokines, we examined the role of SHP-1 in functions of chemokines using viable motheaten (me(v)/me(v)) mice that were deficient in SHP-1. Chemotactic responses to stromal call-derived factor 1 (SDF-1), a CXC chemokine, were enhanced with bone marrow myeloid progenitor cells as well as macrophages, T cells, and B cells from me(v)/me(v) versus wild-type (+/+) mice. SDF-1-dependent actin polymerization and activation of mitogen-activated protein kinases were also greater in me(v)/me(v) versus +/+ cells. In contrast, immature subsets of me(v)/me(v) bone marrow myeloid progenitors were resistant to effects of a number of chemokines that suppressed proliferation of +/+ progenitors. These altered chemokine responses did not appear to be due to enhanced expression of CXCR4 or lack of chemokine receptor expression. However, expression of some chemokine receptors (CCR1, CCR2, CCR3, and CXCR2) was significantly enhanced in me(v)/me(v) T cells. Our results implicate SHP-1 involvement in a number of different chemokine-induced biological activities.
Collapse
Affiliation(s)
- Chang H. Kim
- From the Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Cheng-Kui Qu
- From the Department of Biochemistry/Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Giao Hangoc
- From the Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Scott Cooper
- From the Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Naoyuki Anzai
- From the Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Gen-Sheng Feng
- From the Department of Biochemistry/Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| | - Hal E. Broxmeyer
- From the Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- From the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46208
| |
Collapse
|
21
|
Broxmeyer HE, Kim CH. Regulation of hematopoiesis in a sea of chemokine family members with a plethora of redundant activities. Exp Hematol 1999; 27:1113-23. [PMID: 10390186 DOI: 10.1016/s0301-472x(99)00045-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The field of chemokine biology is a rapidly advancing one, with over 50 chemokines identified that mediate their effects through one or more of 16 different chemokine receptors. Chemokines, originally identified as chemotactic cytokines, manifest a number of functions, including modulation of blood cell production at the level of hematopoietic stem/progenitor cells and the directed movement of these early blood cells. This report reviews chemokines and chemokine/receptor activities mainly in the context of hematopoietic cell regulation and the numerous chemokines that manifest suppressive activity on proliferation of stem/progenitor cells. This is contrasted with the specificity of only a few chemokines for the chemotaxis of these early cells. The large number of chemokines with suppressive activity is hypothesized to reflect the different cell, tissue, and organ sites of production of these chemokines and the need to control stem/progenitor cell proliferation in different organ sites throughout the body.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology/Immunology, Walther Oncology Center, Indiana University School of Medicine, and the Walther Cancer Institute, Indianapolis 46202-5254, USA.
| | | |
Collapse
|
22
|
Gotoh A, Reid S, Miyazawa K, Broxmeyer HE. SDF-1 suppresses cytokine-induced adhesion of human haemopoietic progenitor cells to immobilized fibronectin. Br J Haematol 1999; 106:171-4. [PMID: 10444182 DOI: 10.1046/j.1365-2141.1999.01517.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The process of haemopoietic cell homing to the microenvironment includes migration and adhesion. SDF-1 is a C-X-C chemokine that acts as a chemoattractant for haemopoietic progenitors. Adhesion of haemopoietic progenitors to immobilized fibronectin is up-regulated by stimulation with cytokines. We assessed the effects of SDF-1 on cytokine-induced adhesion of progenitor cells to fibronectin. In factor-dependent human MO7e cells and primary CD34+ cord blood cells, treatment with SDF-1 dose-dependently suppressed cytokine-induced adhesion. Pretreatment with pertussis toxin reversed adhesion-inhibition, suggesting that activation of G-coupled proteins are involved in the intracellular signalling of this process. These data suggest that SDF-1 not only acts as a chemoattractant but also participates in modulating adhesiveness of haemopoietic progenitors to extracellular matrix components.
Collapse
Affiliation(s)
- A Gotoh
- First Department of Internal Medicine, Tokyo Medical University, Tokyo, Japan
| | | | | | | |
Collapse
|
23
|
Abstract
Chemokines regulate hematopoiesis in part by influencing the proliferative status of myeloid progenitor cells (MPC). Human MCP-1/murine JE, a myelosuppressive chemokine, specifically binds C-C chemokine receptor 2 (CCR2). Transgenic mice containing a targeted disruption in CCR2 that prevents expression of CCR2 mRNA and protein and have MPC that are insensitive to inhibition by MCP-1 and JE in vitro were assessed for potential abnormalities in growth of bone marrow (BM) and spleen MPC. MPC in both unseparated and c-kit+lin− populations of BM from CCR2-deficient (−/−) mice were in a greatly increased proliferation state compared with CCR2 littermate control (+/+) mice, an effect not apparent with progenitors from spleens of CCR2 (−/−) mice. Increased cycling status of CCR2 (−/−) BM MPC did not result in increased numbers of nucleated cells or MPC in BM or spleens of CCR2 (−/−) mice. Possible reasons for this apparent discrepancy were highlighted by flow cytometric analysis of c-kit+lin− BM cells and colony formation by MPC subjected to delayed addition of growth factors. The c-kit+lin− population of BM cells from CCR2 (−/−) mice had a significantly higher percentage of apoptotic cells than those from CCR2 (+/+) BM. However, elevated apoptosis was not associated with decreased numbers of c-kit+lin− cells. The increased percentage of apoptotic c-kit+lin− cells was due to elevated apoptosis within the c-kitdimlin−, but not the c-kitbrightlin−, subpopulations of cells. Consistent with enhanced apoptosis of phenotypically defined cells, MPC from CCR2 (−/−) BM and purified c-kit+lin− cells demonstrated decreased cell survival in vitro upon delayed addition of growth factors. The data suggest that signals received by CCR2 limit proliferation of progenitor cells in the BM, but also enhance survival of these cells.
Collapse
|
24
|
Enhanced Myeloid Progenitor Cell Cycling and Apoptosis in Mice Lacking the Chemokine Receptor, CCR2. Blood 1999. [DOI: 10.1182/blood.v93.5.1524] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Chemokines regulate hematopoiesis in part by influencing the proliferative status of myeloid progenitor cells (MPC). Human MCP-1/murine JE, a myelosuppressive chemokine, specifically binds C-C chemokine receptor 2 (CCR2). Transgenic mice containing a targeted disruption in CCR2 that prevents expression of CCR2 mRNA and protein and have MPC that are insensitive to inhibition by MCP-1 and JE in vitro were assessed for potential abnormalities in growth of bone marrow (BM) and spleen MPC. MPC in both unseparated and c-kit+lin− populations of BM from CCR2-deficient (−/−) mice were in a greatly increased proliferation state compared with CCR2 littermate control (+/+) mice, an effect not apparent with progenitors from spleens of CCR2 (−/−) mice. Increased cycling status of CCR2 (−/−) BM MPC did not result in increased numbers of nucleated cells or MPC in BM or spleens of CCR2 (−/−) mice. Possible reasons for this apparent discrepancy were highlighted by flow cytometric analysis of c-kit+lin− BM cells and colony formation by MPC subjected to delayed addition of growth factors. The c-kit+lin− population of BM cells from CCR2 (−/−) mice had a significantly higher percentage of apoptotic cells than those from CCR2 (+/+) BM. However, elevated apoptosis was not associated with decreased numbers of c-kit+lin− cells. The increased percentage of apoptotic c-kit+lin− cells was due to elevated apoptosis within the c-kitdimlin−, but not the c-kitbrightlin−, subpopulations of cells. Consistent with enhanced apoptosis of phenotypically defined cells, MPC from CCR2 (−/−) BM and purified c-kit+lin− cells demonstrated decreased cell survival in vitro upon delayed addition of growth factors. The data suggest that signals received by CCR2 limit proliferation of progenitor cells in the BM, but also enhance survival of these cells.
Collapse
|
25
|
Coreceptor/Chemokine Receptor Expression on Human Hematopoietic Cells: Biological Implications for Human Immunodeficiency Virus–Type 1 Infection. Blood 1999. [DOI: 10.1182/blood.v93.4.1145.404k17_1145_1156] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The recent discovery of chemokine receptors as coreceptors for human immunodeficiency virus–type 1 (HIV-1) entry offers new avenues for investigating the pathogenesis of acquired immunodeficiency syndrome (AIDS)-related cytopenias. To this end, we sought to (1) phenotype human hematopoietic cells for CD4 and the HIV-1 coreceptors CXCR4, CCR5, CCR3, and CCR2b; (2) correlate CD4 and chemokine receptor expression with their susceptibility to HIV-1 infection; and (3) examine any potential interplay between inflammatory cytokines released during HIV-1 infection and regulation of chemokine receptor expression. Fluorescence-activated cell sorting (FACS) analysis of bone marrow mononuclear cells (BMMNC), cells derived from serum-free expanded hematopoietic lineages (colony-forming unit–granulocyte-macrophage [CFU-GM], colony-forming unit-megakaryocyte [CFU-Meg], and burst-forming unit-erythroid [BFU-E]), and CD34+ cells showed differential expression of chemokine receptors and CD4 with some lineage specificity. Significantly, FACS-sorted CXCR4+/CD34+ cells had the same clonogeneic potential as CXCR4−/CD34+ cells. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of FACS-sorted human candidate stem cells (HSC; CD34+, c-kit+, Rho123low) showed the presence of CXCR4 mRNA but not CD4 mRNA. Infection studies with HIV-1 Env-pseudotyped luciferase reporter viruses indicated that X4 Env (CXCR4-using) pseudotypes infected megakaryocytic cells, whereas R5 Env (CCR5-using) pseudotypes did not. Similarly, R5 but not X4 Env-pseudotyped viruses infected granulocyte-macrophage cells in a CD4/CCR5-dependent manner. Erythroid cells were resistant to R5 or X4 viral infection. Finally, we found that γ-interferon treatment upregulated CXCR4 expression on primary hematopoietic cells. In summary, the delineation of chemokine receptor expression on primary hematopoietic cells is a first step towards dissecting the chemokine-chemokine receptor axes that may play a role in hematopoietic cell proliferation and homing. Furthermore, susceptibility of hematopoietic cells to HIV-1 infection is likely to be more complicated than the mere physical presence of CD4 and the cognate chemokine receptor. Lastly, our results suggest a potential interplay between γ-interferon secretion and CXCR4 expression.
Collapse
|
26
|
Coreceptor/Chemokine Receptor Expression on Human Hematopoietic Cells: Biological Implications for Human Immunodeficiency Virus–Type 1 Infection. Blood 1999. [DOI: 10.1182/blood.v93.4.1145] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The recent discovery of chemokine receptors as coreceptors for human immunodeficiency virus–type 1 (HIV-1) entry offers new avenues for investigating the pathogenesis of acquired immunodeficiency syndrome (AIDS)-related cytopenias. To this end, we sought to (1) phenotype human hematopoietic cells for CD4 and the HIV-1 coreceptors CXCR4, CCR5, CCR3, and CCR2b; (2) correlate CD4 and chemokine receptor expression with their susceptibility to HIV-1 infection; and (3) examine any potential interplay between inflammatory cytokines released during HIV-1 infection and regulation of chemokine receptor expression. Fluorescence-activated cell sorting (FACS) analysis of bone marrow mononuclear cells (BMMNC), cells derived from serum-free expanded hematopoietic lineages (colony-forming unit–granulocyte-macrophage [CFU-GM], colony-forming unit-megakaryocyte [CFU-Meg], and burst-forming unit-erythroid [BFU-E]), and CD34+ cells showed differential expression of chemokine receptors and CD4 with some lineage specificity. Significantly, FACS-sorted CXCR4+/CD34+ cells had the same clonogeneic potential as CXCR4−/CD34+ cells. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of FACS-sorted human candidate stem cells (HSC; CD34+, c-kit+, Rho123low) showed the presence of CXCR4 mRNA but not CD4 mRNA. Infection studies with HIV-1 Env-pseudotyped luciferase reporter viruses indicated that X4 Env (CXCR4-using) pseudotypes infected megakaryocytic cells, whereas R5 Env (CCR5-using) pseudotypes did not. Similarly, R5 but not X4 Env-pseudotyped viruses infected granulocyte-macrophage cells in a CD4/CCR5-dependent manner. Erythroid cells were resistant to R5 or X4 viral infection. Finally, we found that γ-interferon treatment upregulated CXCR4 expression on primary hematopoietic cells. In summary, the delineation of chemokine receptor expression on primary hematopoietic cells is a first step towards dissecting the chemokine-chemokine receptor axes that may play a role in hematopoietic cell proliferation and homing. Furthermore, susceptibility of hematopoietic cells to HIV-1 infection is likely to be more complicated than the mere physical presence of CD4 and the cognate chemokine receptor. Lastly, our results suggest a potential interplay between γ-interferon secretion and CXCR4 expression.
Collapse
|
27
|
Abstract
Over the past decade, with the advent of hematopoietic growth factors, major strides have been made and multiple studies have attempted to define the use of these cytokines in acute leukemia. It is perhaps disappointing that, after so many studies, so many questions remain. Nevertheless, the role of cytokines in induction therapy seems to be established, although questions remain around the issue of priming therapy. Intriguing data regarding the potential for enhancing antimicrobial function should hopefully be resolved over the next few years. What is perhaps most reassuring is that the issue of safety, which for a considerable period of time precluded the development of clinical trials in acute leukemia, has been firmly laid to rest. The use of growth factors to protect normal stem cells during treatment of leukemia and to induce leukemic cell differentiation has not yet been the subject of many clinical trials. Also, growth factors are likely targets for the interruption of autocrine leukemic blast or progenitor cell growth, but again, few clinical observations are published. With the ongoing cloning of new growth factors active both in normal hematopoiesis and in leukemogenesis, the role of growth factor use in the treatment of AML will likely be the basis for much future preclinical and clinical activity.
Collapse
Affiliation(s)
- J M Rowe
- Hematology Institute, Rambam Medical Ce2nter, Hifa, Israel
| | | |
Collapse
|
28
|
Soto H, Wang W, Strieter RM, Copeland NG, Gilbert DJ, Jenkins NA, Hedrick J, Zlotnik A. The CC chemokine 6Ckine binds the CXC chemokine receptor CXCR3. Proc Natl Acad Sci U S A 1998; 95:8205-10. [PMID: 9653165 PMCID: PMC20954 DOI: 10.1073/pnas.95.14.8205] [Citation(s) in RCA: 187] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/1998] [Indexed: 02/08/2023] Open
Abstract
We cloned the mouse homologue of the chemokine receptor CXCR3, which is located in mouse chromosome X. We screened a large panel of chemokines for their ability to induce a calcium flux in mouse CXCR3-transfected cells and identified a new ligand for this receptor, the recently reported CC chemokine 6Ckine. This represents an example of a CC chemokine, which binds to a CXC chemokine receptor. Like other ligands of this receptor, 6Ckine has angiostatic properties. 6Ckine is known to chemoattract T cells. In line with this, CXCR3 is expressed preferentially in Th1 cells and in lymphoid organs of the IL-10(-/-) mouse that develops chronic colitis. Its ability to attract T cells as well as its angiostatic properties suggest that 6Ckine may be an effective anti-tumor agent.
Collapse
Affiliation(s)
- H Soto
- DNAX Research Institute for Cellular and Molecular Biology, Palo Alto, CA 94304, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Characterization of CKβ8 and CKβ8-1: Two Alternatively Spliced Forms of Human β-Chemokine, Chemoattractants for Neutrophils, Monocytes, and Lymphocytes, and Potent Agonists at CC Chemokine Receptor 1. Blood 1998. [DOI: 10.1182/blood.v91.9.3118.3118_3118_3126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Two new members of human β-chemokine cDNA were isolated based on structural and functional similarities to human leukotactin-1. One of these clones was identical to the previously isolated human β-chemokine, CKβ8, whereas the other is a splicing variant of CKβ8, therefore named CKβ8-1. CKβ8 was short in 51 nucleotides (17 amino acids) compared with CKβ8-1. The mature proteins of CKβ8-1 and CKβ8 consisted of 116 and 99 amino acids with calculated molecular weights of 12,500 and 10,950, respectively. Both CKβ8-1 and CKβ8 were potent agonists at CCR1. These chemokines chemoattracted neutrophils, monocytes, and lymphocytes. They also significantly suppressed colony formation by human bone marrow, granulocyte-macrophage, erythroid, and multipotential progenitor cells stimulated by combinations of growth factors. To our knowledge, this is the first example that an alternative splicing produces two active β-chemokines from a single gene.
Collapse
|
30
|
Characterization of CKβ8 and CKβ8-1: Two Alternatively Spliced Forms of Human β-Chemokine, Chemoattractants for Neutrophils, Monocytes, and Lymphocytes, and Potent Agonists at CC Chemokine Receptor 1. Blood 1998. [DOI: 10.1182/blood.v91.9.3118] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Two new members of human β-chemokine cDNA were isolated based on structural and functional similarities to human leukotactin-1. One of these clones was identical to the previously isolated human β-chemokine, CKβ8, whereas the other is a splicing variant of CKβ8, therefore named CKβ8-1. CKβ8 was short in 51 nucleotides (17 amino acids) compared with CKβ8-1. The mature proteins of CKβ8-1 and CKβ8 consisted of 116 and 99 amino acids with calculated molecular weights of 12,500 and 10,950, respectively. Both CKβ8-1 and CKβ8 were potent agonists at CCR1. These chemokines chemoattracted neutrophils, monocytes, and lymphocytes. They also significantly suppressed colony formation by human bone marrow, granulocyte-macrophage, erythroid, and multipotential progenitor cells stimulated by combinations of growth factors. To our knowledge, this is the first example that an alternative splicing produces two active β-chemokines from a single gene.
Collapse
|
31
|
Daliani D, Ulmer RA, Jackow C, Pugh W, Gansbacher B, Cabanillas F, Duvic M, Sarris AH. Tumor necrosis factor-alpha and interferon-gamma, but not HTLV-I tax, are likely factors in the epidermotropism of cutaneous T-cell lymphoma via induction of interferon-inducible protein-10. Leuk Lymphoma 1998; 29:315-28. [PMID: 9684929 DOI: 10.3109/10428199809068568] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have previously shown that Interferon-Inducible Protein-10 (IP-10), a cytokine chemotactic for CD4-positive lymphocytes, is overexpressed by lesional epidermal keratinocytes and probably accounts for the epidermotropism of cutaneous T-cell lymphoma (CTCL). The tax gene of human T-lymphotropic virus-I (HTLV-I) immortalizes CD4-positive lymphocytes, induces IFN-gamma, and has been detected in patients with classical CTCL who are seronegative for HTLV-I. TNF-alpha is synergistic with IFN-gamma for the induction of IP-10. We therefore decided to define the presence of tax, IFN-gamma, TNF-alpha, and IP-10 in lesions of 19 adults with classical CTCL who were seronegative for HTLV-I. Lesional mRNAs for actin, TNF-alpha, IFN-gamma, and tax were detected by reverse-transcriptase polymerase chain reaction (RT-PCR) amplification. In addition IP-10, TNF-alpha, and IFN-gamma were detected and localized with immunocytochemistry of frozen sections. In agreement with previous observations IP-10 was overexpressed in lesional keratinocytes of all 19 patients. By RT-PCR, mRNA for IFN-gamma was detected in lesions of 8, and for TNF-alpha in lesions of 13 patients. By immunocytochemistry, TNF-alpha was expressed by lesional keratinocytes in 10 of 13 tested patients, whereas IFN-gamma was focally expressed by lesional lymphocytes and faintly by lesional keratinocytes in 9 of 13 tested patients. tax mRNA was not detected in lesions of any patient, but was easily detectable in cutaneous lesions or peripheral blood of control patients who were seropositive for HTLV-I. We conclude that TNF-alpha and IFN-gamma may cause epidermotropism by inducing IP-10. However, the tax gene of HTLV-I does not appear to be involved in the pathogenesis of classical CTCL.
Collapse
Affiliation(s)
- D Daliani
- Department of Hematology, The University of Texas M.D. Anderson Cancer Center, Houston, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Broxmeyer HE, Orazi A, Hague NL, Sledge GW, Rasmussen H, Gordon MS. Myeloid progenitor cell proliferation and mobilization effects of BB10010, a genetically engineered variant of human macrophage inflammatory protein-1alpha, in a phase I clinical trial in patients with relapsed/refractory breast cancer. Blood Cells Mol Dis 1998; 24:14-30. [PMID: 9516378 DOI: 10.1006/bcmd.1998.0167] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Macrophage Inflammatory Protein (MIP)-1alpha is myelosuppressive in vitro and in vivo for hematopoietic stem and immature subsets of myeloid progenitor cells, demonstrates some myeloprotective effects in mice treated with Ara-C and hydroxyurea, and has stem/progenitor cell mobilizing activity in mice. Based on these observations, BB10010, a genetic variant of MIP-1alpha, was assessed for effects on marrow and blood myeloid progenitor cells in patients with relapsed/refractory breast cancer. MIP-1alpha readily polymerizes, whereas BB10010 has a reduced tendency to form large polymers at physiological pH and ionic strength and retains biological activity. Patients were injected with 5, 10, 30 or 100 microg/kg BB10010 s.c. daily for 3 days. BB10010 significantly reduced the cycling status of marrow myeloid progenitors from pretreatment levels of 39-58% to 0 - 11% one day after the third and last injection of BB10010. This was associated with significant decreases in frequency of marrow progenitors (number of colonies formed per number of cells plated) and percent biopsied marrow CD34+ cells. The suppressive effects were reversible in patients and the rapidity of this reversal demonstrated in mouse studies. BB10010 had no effect on nucleated cellularity or on the proliferation of nucleated cells as assessed in marrow biopsies from the patients. These latter effects may in part reflect the noted decreased apoptosis of nucleated cells by BB10010. BB10010 also demonstrated significant but modest myeloid progenitor cell mobilizing capacity. Blood progenitors were in a slow or non-cycling state prior to treatment and this did not change after administration of BB10010. The above effects of BB10010 were similar at the four different dosage levels assessed. These results demonstrate in humans the suppressive and mobilizing effects of MIP-1alpha and BB10010 previously noted in vivo in mice.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology/Immunology and Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | |
Collapse
|
33
|
McFadden G, Kelvin D. New strategies for chemokine inhibition and modulation: you take the high road and I'll take the low road. Biochem Pharmacol 1997; 54:1271-80. [PMID: 9393669 DOI: 10.1016/s0006-2952(97)00182-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemokines are low molecular weight cytokines that induce extravasation, chemotaxis, and activation of a wide variety of leukocytes. Members of the different chemokine families are defined by the orientation of specific critical cysteine residues, and are designated as C-X-C (e.g. interleukin-8), C-C (e.g. regulated upon activation normally T cell expressed and secreted, RANTES), or C (lymphotactin). All chemokines bind to members of a G-protein coupled serpentine receptor superfamily that span the leukocyte cell surface membrane seven times and mediate the biological activities of the individual ligands. Most chemokines possess two major binding surfaces: a high affinity site responsible for specific ligand/receptor interactions and a lower affinity site, also called the heparin-binding or glycosaminoglycan-binding domain, believed to be responsible for the establishment and presentation of chemokine gradients on the surface of endothelial cells and within the extracellular matrix. Although chemokines are clearly beneficial in wound healing, hemopoiesis, and the clearance of infectious organisms, the continued expression of chemokines is associated with chronic inflammation. Therefore, this class of cytokines are attractive targets for the creation of antagonists that abrogate one or more chemokine functions. It is envisioned that such antagonists could serve as a new class of anti-inflammatory drugs. In this commentary, we will discuss two different but related strategies for antagonizing chemokine-induced functions, namely, disruption of the low and high affinity binding sites.
Collapse
Affiliation(s)
- G McFadden
- Department of Microbiology and Immunology, University of Western Ontario, London, Canada.
| | | |
Collapse
|
34
|
Boring L, Gosling J, Chensue SW, Kunkel SL, Farese RV, Broxmeyer HE, Charo IF. Impaired monocyte migration and reduced type 1 (Th1) cytokine responses in C-C chemokine receptor 2 knockout mice. J Clin Invest 1997; 100:2552-61. [PMID: 9366570 PMCID: PMC508456 DOI: 10.1172/jci119798] [Citation(s) in RCA: 860] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) is a potent agonist for mononuclear leukocytes and has been implicated in the pathogenesis of atherosclerosis and granulomatous lung disease. To determine the role of MCP-1 and related family members in vivo, we used homologous recombination in embryonic stem cells to generate mice with a targeted disruption of C-C chemokine receptor 2 (CCR2), the receptor for MCP-1. CCR2-/- mice were born at the expected Mendelian ratios and developed normally. In response to thioglycollate, the recruitment of peritoneal macrophages decreased selectively. In in vitro chemotaxis assays, CCR2-/- leukocytes failed to migrate in response to MCP-1. Granulomatous lung disease was induced in presensitized mice by embolization with beads coupled to purified protein derivative (PPD) of Mycobacterium bovis. As compared with wild-type littermates, CCR2-/- mice had a decrease in granuloma size accompanied by a dramatic decrease in the level of interferon gamma in the draining lymph nodes. Production of interferon gamma was also decreased in PPD-sensitized splenocytes from CCR2-/- mice and in naive splenocytes activated by concanavalin A. We conclude that CCR2-/- mice have significant defects in both delayed-type hypersensitivity responses and production of Th1-type cytokines. These data suggest an important and unexpected role for CCR2 activation in modulating the immune response, as well as in recruiting monocytes/macrophages to sites of inflammation.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/cytology
- Chemokine CCL2/pharmacology
- Chemokines/pharmacology
- Chemotaxis, Leukocyte/genetics
- Chemotaxis, Leukocyte/physiology
- Cytokines/biosynthesis
- Embryo, Mammalian
- Granuloma, Respiratory Tract/immunology
- Granuloma, Respiratory Tract/microbiology
- Granuloma, Respiratory Tract/physiopathology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/physiology
- Humans
- Lung Diseases/immunology
- Lung Diseases/physiopathology
- Lymph Nodes/immunology
- Macrophages, Alveolar/immunology
- Mice
- Mice, Knockout
- Monocytes/physiology
- Mycobacterium bovis
- Receptors, CCR2
- Receptors, CCR5/biosynthesis
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/physiology
- Recombinant Proteins/pharmacology
- Th1 Cells/immunology
- Transcription, Genetic
- Tuberculin
Collapse
Affiliation(s)
- L Boring
- Gladstone Institute of Cardiovascular Disease, University of California, San Francisco 94141-9100, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Sanchez X, Cousins-Hodges B, Aguilar T, Gosselink P, Lu Z, Navarro J. Activation of HIV-1 coreceptor (CXCR4) mediates myelosuppression. J Biol Chem 1997; 272:27529-31. [PMID: 9346884 DOI: 10.1074/jbc.272.44.27529] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chemokines are cytokines that activate and induce the migration of leukocytes. Stroma-derived factor-1 (SDF-1) is a novel chemokine that blocks the entry of T-tropic HIV-1 mediated by fusin/CXCR4/LESTR (leukocyte-derived seven-transmembrane domain receptor). In this work we demonstrate that SDF-1 triggers increases in intracellular calcium and inhibits the proliferation of myeloid progenitor cell line 32D. By contrast, SDF-1 neither triggers a calcium response nor affects the proliferation of the myeloid progenitor cell line 32D-GR that is deficient in CXCR4. Responsiveness to SDF-1 was rescued by transfection of 32D-GR cells with a cDNA encoding the human CXCR4. The data indicate that SDF-1 induces myelosuppression by activation of CXCR4. The constitutive production of SDF-1 by bone marrow stromal cells argues for a major role of SDF-1 on the regulation of myelopoiesis.
Collapse
Affiliation(s)
- X Sanchez
- Department of Physiology & Biophysics and Sealy Center for Molecular Science, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | | | |
Collapse
|
36
|
Curfs JH, Meis JF, Hoogkamp-Korstanje JA. A primer on cytokines: sources, receptors, effects, and inducers. Clin Microbiol Rev 1997; 10:742-80. [PMID: 9336671 PMCID: PMC172943 DOI: 10.1128/cmr.10.4.742] [Citation(s) in RCA: 200] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Protection against pathogens is a prerequisite for survival of most organisms. To cope with this continuous challenge, complex defense mechanisms have evolved. The construction, adaptation, and maintenance of these mechanisms are under control of an extensive network of regulatory proteins called cytokines. A great number of cytokines have been described over the last 2 decades. This review consists of an overview of cytokines that are involved in immune responses and describes some historical and general aspects as well as prospective clinical applications. Major biological effects together with information on cytokine receptors, producers, inducers, and biochemical and molecular characteristics are listed in tables. In addition, some basic information is given on cytokine receptor signal transduction. Finally, the recent discoveries of cytokine receptors functioning as coreceptors in the pathogenesis of human immunodeficiency virus are summarized.
Collapse
Affiliation(s)
- J H Curfs
- Department of Medical Microbiology, University Hospital Nijmegen, The Netherlands.
| | | | | |
Collapse
|
37
|
Krathwohl MD, Hromas R, Brown DR, Broxmeyer HE, Fife KH. Functional characterization of the C---C chemokine-like molecules encoded by molluscum contagiosum virus types 1 and 2. Proc Natl Acad Sci U S A 1997; 94:9875-80. [PMID: 9275219 PMCID: PMC23285 DOI: 10.1073/pnas.94.18.9875] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Many viruses have evolved mechanisms for evading the host immune system by synthesizing proteins that interfere with the normal immune response. The poxviruses are among the most accomplished at deceiving their hosts' immune systems. The nucleotide sequence of the genome of the human cutaneous poxvirus, molluscum contagiosum virus (MCV) type 1, was recently reported to contain a region that resembles a human chemokine. We have cloned and expressed the chemokine-like genes from MCV type 1 and the closely related MCV type 2 to determine a potential role for these proteins in the viral life cycle. In monocyte chemotaxis assays, the viral proteins have no chemotactic activity but both viral proteins block the chemotactic response to the human chemokine, macrophage inflammatory protein (MIP)-1alpha. Like MIP-1alpha, both viral proteins also inhibit the growth of human hematopoietic progenitor cells, but the viral proteins are more potent in this activity than the human chemokine. These viral chemokines antagonize the chemotactic activity of human chemokines and have an inhibitory effect on human hematopoietic progenitor cells. We hypothesize that the inhibition of chemotaxis is an immune evasion function of these proteins during molluscum contagiosum virus infection. The significance of hematopoietic progenitor cell inhibition in viral pathogenesis is uncertain.
Collapse
Affiliation(s)
- M D Krathwohl
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
38
|
Coleman TA, Parmelee D, Thotakura NR, Nguyen N, Bürgin M, Gentz S, Gentz R. Production and purification of novel secreted human proteins. Gene 1997; 190:163-71. [PMID: 9185863 DOI: 10.1016/s0378-1119(96)00711-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Our objective during the last year was to produce and purify 50-80 novel, secreted human proteins identified via high throughput cDNA sequencing and computer analysis. We chose the baculovirus expression vector system in order to obtain secreted, correctly folded, bioactive proteins. Recombinant (re-)baculoviruses (BV) were plaque purified, and pulse-labeling was used to verify the synthesis and secretion of the re-proteins. N-terminal microsequencing was performed to simultaneously confirm the identity of the protein(s) as well as the signal peptide (SP) cleavage site(s). Following sequence confirmation, the proteins were purified to homogeneity and functional assays carried out to determine potential therapeutic applications. We identified proteins with antiviral activity, several novel growth factors, proteins influencing the differentiation of specific cell types, novel proteases and protease inhibitors among others. Certain proteins were expressed both in insect cells and in CHO stable cell lines. In the cases analyzed, we found that the same SP cleavage site was utilized in the two expression systems. Significant differences were observed in the carbohydrate moieties attached to the proteins, though no effects on the biological activity due to these differences have been demonstrated. The BV system has served as a viable alternative for the high throughput, high fidelity expression of many novel secreted human genes. To date, more than 75 new genes have been expressed, and the re-proteins purified. This expression system combines many favorable traits including relative speed, moderate cost but perhaps most importantly, the production of biologically active proteins.
Collapse
Affiliation(s)
- T A Coleman
- Human Genome Sciences Inc., Rockville, MD 20850-3338, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Hieshima K, Imai T, Opdenakker G, Van Damme J, Kusuda J, Tei H, Sakaki Y, Takatsuki K, Miura R, Yoshie O, Nomiyama H. Molecular cloning of a novel human CC chemokine liver and activation-regulated chemokine (LARC) expressed in liver. Chemotactic activity for lymphocytes and gene localization on chromosome 2. J Biol Chem 1997; 272:5846-53. [PMID: 9038201 DOI: 10.1074/jbc.272.9.5846] [Citation(s) in RCA: 289] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Partial overlapping cDNA sequences likely to encode a novel human CC chemokine were identified from the GenBank Expressed Sequence Tag data base. Using these sequences, we isolated full-length cDNA encoding a protein of 96 amino acid residues with 20-28% identity to other CC chemokines. By Northern blot, this chemokine was mainly expressed in liver among various tissues and strongly induced in several human cell lines by phorbol myristate acetate. We thus designated this chemokine as LARC from Liver and Activation-Regulated Chemokine. We mapped the LARC gene close to the chromosomal marker D2S159 at chromosome 2q33-q37 by somatic cell and radiation hybrid mappings and isolated two yeast artificial chromosome clones containing the LARC gene from this region. To prepare LARC, we subcloned the cDNA into a baculovirus vector and expressed it in insect cells. The secreted protein started at Ala-27 and was significantly chemotactic for lymphocytes. At a concentration of 1 microg/ml, it also showed a weak chemotactic activity for granulocytes. Unlike other CC chemokines, however, LARC was not chemotactic for monocytic THP-1 cells or blood monocytes. LARC tagged with secreted alkaline phosphatase-(His)6 bound specifically to lymphocytes, the binding being competed only by LARC and not by other CC or CXC chemokines. Scatchard analysis revealed a single class of receptors for LARC on lymphocytes with a Kd of 0.4 nM and 2100 sites/cell. Collectively, LARC is a novel CC chemokine, which may represent a new group of CC chemokines localized on chromosome 2.
Collapse
Affiliation(s)
- K Hieshima
- Department of Biochemistry, Kumamoto University Medical School, Honjo, Kumamoto 860, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Broxmeyer HE, Cooper S, Cacalano G, Hague NL, Bailish E, Moore MW. Involvement of Interleukin (IL) 8 receptor in negative regulation of myeloid progenitor cells in vivo: evidence from mice lacking the murine IL-8 receptor homologue. J Exp Med 1996; 184:1825-32. [PMID: 8920870 PMCID: PMC2192871 DOI: 10.1084/jem.184.5.1825] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Expansion of mature neutrophils has been observed in mice lacking the murine interleukin (IL) 8 receptor homolog [mIL-8Rh(-/-)], and human (hu) IL-8 suppresses proliferation of primitive myeloid cells in vitro and in vivo. To evaluate involvement and relevance of murine IL-8 receptor homolog (mIL-8Rh) in negative regulation of myelopoiesis, we studied mIL-8Rh(-/-) and (+/+) mice raised in a normal or germ-free environment. Immature myeloid progenitors from mIL-8Rh(+/+) mice bred under normal or germ-free conditions were significantly suppressed in vitro by recombinant huIL-8, macrophage inflammatory protein (MIP)-1 alpha, platelet factor (PF) 4, interferon inducible protein (IP) 10, monocyte chemotactic peptide (MCP) 1, and H-ferritin. In contrast, progenitors from mIL-8Rh(-/-) mice were insensitive to inhibition by IL-8, but not to these other chemokines and H-ferritin. Mouse MIP-2, a ligand for mIL-8Rh, suppressed progenitors from normal but not mIL-8Rh(-/-) mice. Under normal environmental conditions, enhanced numbers of myeloid progenitors were found in femur, spleen, and blood of mIL-8Rh(-/-) compared with mIL-8Rh(+/+) mice. Numbers of myeloid progenitors were greatly decreased in mIL-8Rh(-/-)and (+/+) mice in germ-free conditions, and were either not significantly enhanced in mIL-8Rh(-/-) mice compared with (+/+) mice or were only moderately so. Differences in progenitors/organ between a germ-free and normal environment were greater for the mIL-8Rh(-/-) mice. These results document selective insensitivity of myeloid progenitor cells from mIL-8Rh(-/-) mice to inhibition by huIL-8 and mouse MIP-2 and a large expansion of myeloid progenitors in these mice, the latter effect being environmentally inducible. This provides strong support for a negative myeloid regulatory role played by the mIL-8Rh in vivo, whose active ligand may be MIP-2.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Medicine (Hematology/Oncology), Indiana University School of Medicine, Indianapolis 46202, USA
| | | | | | | | | | | |
Collapse
|
41
|
Broxmeyer HE. Is interleukin 17, an inducible cytokine that stimulates production of other cytokines, merely a redundant player in a sea of other biomolecules? J Exp Med 1996; 183:2411-5. [PMID: 8676060 PMCID: PMC2192608 DOI: 10.1084/jem.183.6.2411] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- H E Broxmeyer
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
42
|
CORD BLOOD STEM AND PROGENITOR CELL THERAPY FOR IMMUNODEFICIENCY AND OTHER DISORDERS. Immunol Allergy Clin North Am 1996. [DOI: 10.1016/s0889-8561(05)70255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
43
|
Broxmeyer HE. CORD BLOOD STEM AND PROGENITOR CELL THERAPY FOR IMMUNODEFICIENCY AND OTHER DISORDERS. Radiol Clin North Am 1996. [DOI: 10.1016/s0033-8389(22)00220-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
|