1
|
Minor BMN, LeMoine D, Seger C, Gibbons E, Koudouovoh J, Taya M, Kurtz D, Xu Y, Hammes SR. Estradiol Augments Tumor-Induced Neutrophil Production to Promote Tumor Cell Actions in Lymphangioleiomyomatosis Models. Endocrinology 2023; 164:bqad061. [PMID: 37042477 PMCID: PMC10164661 DOI: 10.1210/endocr/bqad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/13/2023]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare cystic lung disease caused by smooth muscle cell-like tumors containing tuberous sclerosis (TSC) gene mutations and found almost exclusively in females. Patient studies suggest LAM progression is estrogen dependent, an observation supported by in vivo mouse models. However, in vitro data using TSC-null cell lines demonstrate modest estradiol (E2) responses, suggesting E2 effects in vivo may involve pathways independent of direct tumor stimulation. We previously reported tumor-dependent neutrophil expansion and promotion of TSC2-null tumor growth in an E2-sensitive LAM mouse model. We therefore hypothesized that E2 stimulates tumor growth in part by promoting neutrophil production. Here we report that E2-enhanced lung colonization of TSC2-null cells is indeed dependent on neutrophils. We demonstrate that E2 induces granulopoiesis via estrogen receptor α in male and female bone marrow cultures. With our novel TSC2-null mouse myometrial cell line, we show that factors released from these cells drive E2-sensitive neutrophil production. Last, we analyzed single-cell RNA sequencing data from LAM patients and demonstrate the presence of tumor-activated neutrophils. Our data suggest a powerful positive feedback loop whereby E2 and tumor factors induce neutrophil expansion, which in turn intensifies tumor growth and production of neutrophil-stimulating factors, resulting in continued TSC2-null tumor growth.
Collapse
Affiliation(s)
- Briaunna M N Minor
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dana LeMoine
- Division of Comparative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Christina Seger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Erin Gibbons
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jules Koudouovoh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Manisha Taya
- Division of Hematology and Oncology, Department of Internal Medicine, UTSW Medical Center, Dallas, TX 75390, USA
| | - Daniel Kurtz
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Yan Xu
- Divisions of Pulmonary Biology & Biomedical Informatics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Stephen R Hammes
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
2
|
Elsehly WM, Mourad GM, Mehanna RA, Kholief MA, El-Nikhely NA, Awaad AK, Attia MH. The potential implications of estrogenic and antioxidant-dependent activities of high doses of methyl paraben on MCF7 breast cancer cells. J Biochem Mol Toxicol 2022; 36:e23012. [PMID: 35174924 DOI: 10.1002/jbt.23012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/05/2021] [Accepted: 01/05/2022] [Indexed: 02/05/2023]
Abstract
Methyl paraben (MP) is an endocrine-disrupting compound that possesses estrogenic properties and contributes to an aberrant burden of estrogen signaling in the human breast and subsequently increasing the risks for the development of breast cancer. The exact exposure, as well as the safe concentrations, are variable among daily products. The present study addresses the effects of exposure to escalated concentrations of MP on the proliferation of MCF-7 breast cancer cells in addition to exploring its other mechanisms of action. The study involved exposure of cultured MCF-7 breast cancer cells to seven MP concentrations, ranging from 40 to 800 µM for 5 days. Cell viability, apoptosis, and proliferation were respectively assessed using crystal violet test, flow cytometric analysis, and quantitative real-time polymerase chain reaction for Ki-67 expression. The estradiol (E2) secretion and oxidative stress were also assessed and analyzed in correlation to MP's proliferation and cytotoxicity potentials. The results showed that the maximum proliferative concentration of MP was 800 µM. At a concentration of 40 μM and higher, MP induced increased expression of Ki-67, denoting enhanced proliferation of the cells in monolayer culture. A positive correlation between the detrimental oxidative stress effect of MP's tested concentrations, cell proliferation, and viability was demonstrated (p < 0.05). Our results indicated that MP at high doses induced sustained cell proliferation due to E2 secretion as well as its antioxidant activity. Accordingly, it was concluded that high and unpredicted exposure to MP might carry a carcinogenic hazard on estrogen receptor-positive breast cancer cells.
Collapse
Affiliation(s)
- Wafaa M Elsehly
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ghada M Mourad
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Department of Histology and Cell Biology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Radwa A Mehanna
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa A Kholief
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nefertiti A El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Ashraf K Awaad
- Center of Excellence for Research in Regenerative Medicine and Applications (CERRMA), Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - MennattAllah H Attia
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Viny AD, Bowman RL, Liu Y, Lavallée VP, Eisman SE, Xiao W, Durham BH, Navitski A, Park J, Braunstein S, Alija B, Karzai A, Csete IS, Witkin M, Azizi E, Baslan T, Ott CJ, Pe'er D, Dekker J, Koche R, Levine RL. Cohesin Members Stag1 and Stag2 Display Distinct Roles in Chromatin Accessibility and Topological Control of HSC Self-Renewal and Differentiation. Cell Stem Cell 2019; 25:682-696.e8. [PMID: 31495782 DOI: 10.1016/j.stem.2019.08.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 06/19/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022]
Abstract
Transcriptional regulators, including the cohesin complex member STAG2, are recurrently mutated in cancer. The role of STAG2 in gene regulation, hematopoiesis, and tumor suppression remains unresolved. We show that Stag2 deletion in hematopoietic stem and progenitor cells (HSPCs) results in altered hematopoietic function, increased self-renewal, and impaired differentiation. Chromatin immunoprecipitation (ChIP) sequencing revealed that, although Stag2 and Stag1 bind a shared set of genomic loci, a component of Stag2 binding sites is unoccupied by Stag1, even in Stag2-deficient HSPCs. Although concurrent loss of Stag2 and Stag1 abrogated hematopoiesis, Stag2 loss alone decreased chromatin accessibility and transcription of lineage-specification genes, including Ebf1 and Pax5, leading to increased self-renewal and reduced HSPC commitment to the B cell lineage. Our data illustrate a role for Stag2 in transformation and transcriptional dysregulation distinct from its shared role with Stag1 in chromosomal segregation.
Collapse
Affiliation(s)
- Aaron D Viny
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Robert L Bowman
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu Liu
- Program in Systems Biology, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Vincent-Philippe Lavallée
- Center for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shira E Eisman
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wenbin Xiao
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Benjamin H Durham
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anastasia Navitski
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jane Park
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephanie Braunstein
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Besmira Alija
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abdul Karzai
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isabelle S Csete
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew Witkin
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elham Azizi
- Center for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Timour Baslan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher J Ott
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Dana Pe'er
- Center for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Job Dekker
- Program in Systems Biology, Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA
| | - Richard Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ross L Levine
- Human Oncology and Pathogenesis Program and Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
6
|
Sugino N, Kawahara M, Tatsumi G, Kanai A, Matsui H, Yamamoto R, Nagai Y, Fujii S, Shimazu Y, Hishizawa M, Inaba T, Andoh A, Suzuki T, Takaori-Kondo A. A novel LSD1 inhibitor NCD38 ameliorates MDS-related leukemia with complex karyotype by attenuating leukemia programs via activating super-enhancers. Leukemia 2017; 31:2303-2314. [PMID: 28210006 DOI: 10.1038/leu.2017.59] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 01/22/2017] [Accepted: 02/03/2017] [Indexed: 12/15/2022]
Abstract
Lysine-specific demethylase 1 (LSD1) regulates gene expression by affecting histone modifications and is a promising target for acute myeloid leukemia (AML) with specific genetic abnormalities. Novel LSD1 inhibitors, NCD25 and NCD38, inhibited growth of MLL-AF9 leukemia as well as erythroleukemia, megakaryoblastic leukemia and myelodysplastic syndromes (MDSs) overt leukemia cells in the concentration range that normal hematopoiesis was spared. NCD25 and NCD38 invoked the myeloid development programs, hindered the MDS and AML oncogenic programs, and commonly upregulated 62 genes in several leukemia cells. NCD38 elevated H3K27ac level on enhancers of these LSD1 signature genes and newly activated ~500 super-enhancers. Upregulated genes with super-enhancer activation in erythroleukemia cells were enriched in leukocyte differentiation. Eleven genes including GFI1 and ERG, but not CEBPA, were identified as the LSD1 signature with super-enhancer activation. Super-enhancers of these genes were activated prior to induction of the transcripts and myeloid differentiation. Depletion of GFI1 attenuated myeloid differentiation by NCD38. Finally, a single administration of NCD38 causes the in vivo eradication of primary MDS-related leukemia cells with a complex karyotype. Together, NCD38 derepresses super-enhancers of hematopoietic regulators that are silenced abnormally by LSD1, attenuates leukemogenic programs and consequently exerts anti-leukemic effect against MDS-related leukemia with adverse outcome.
Collapse
Affiliation(s)
- N Sugino
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - M Kawahara
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - G Tatsumi
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - A Kanai
- Department of Molecular Oncology and Leukemia Program Project, Hiroshima University, Hiroshima, Japan
| | - H Matsui
- Department of Molecular Oncology and Leukemia Program Project, Hiroshima University, Hiroshima, Japan.,Department of Molecular Laboratory Medicine, Kumamoto University, Kumamoto, Japan
| | - R Yamamoto
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Y Nagai
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - S Fujii
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Y Shimazu
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - M Hishizawa
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Inaba
- Department of Molecular Oncology and Leukemia Program Project, Hiroshima University, Hiroshima, Japan
| | - A Andoh
- Department of Medicine, Shiga University of Medical Science, Otsu, Japan
| | - T Suzuki
- Department of Chemistry, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan.,CREST, Japan Science and Technology Agency (JST), Tokyo, Japan
| | - A Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
7
|
Yu X, Wang Y, Lin J, Hu Y, Kawai T, Taubman MA, Han X. Lipopolysaccharides-Induced Suppression of Innate-Like B Cell Apoptosis Is Enhanced by CpG Oligodeoxynucleotide and Requires Toll-Like Receptors 2 and 4. PLoS One 2016; 11:e0165862. [PMID: 27812176 PMCID: PMC5094738 DOI: 10.1371/journal.pone.0165862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/19/2016] [Indexed: 12/30/2022] Open
Abstract
Innate-like B lymphocytes play an important role in innate immunity in periodontal disease through Toll-like receptor (TLR) signaling. However, it is unknown how innate-like B cell apoptosis is affected by the periodontal infection-associated innate signals. This study is to determine the effects of two major TLR ligands, lipopolysaccharide (LPS) and CpG-oligodeoxynucleotides (CpG-ODN), on innate-like B cell apoptosis. Spleen B cells were isolated from wild type (WT), TLR2 knockout (KO) and TLR4 KO mice and cultured with E. coli LPS alone, P. gingivalis LPS alone, or combined with CpG-ODN for 2 days. B cell apoptosis and expressions of specific apoptosis-related genes were analyzed by flow cytometry and real-time PCR respectively. P. gingivalis LPS, but not E. coli LPS, reduced the percentage of AnnexinV+/7-AAD- cells within IgMhighCD23lowCD43-CD93- marginal zone (MZ) B cell sub-population and IgMhighCD23lowCD43+CD93+ innate response activator (IRA) B cell sub-population in WT but not TLR2KO or TLR4KO mice. CpG-ODN combined with P. gingivalis LPS further reduced the percentage of AnnexinV+/7-AAD- cells within MZ B cells and IRA B cells in WT but not TLR2 KO or TLR4 KO mice. Pro-apoptotic CASP4, CASP9 and Dapk1 were significantly down-regulated in P. gingivalis LPS- and CpG-ODN-treated B cells from WT but not TLR2 KO or TLR4 KO mice. Anti-apoptotic IL-10 was significantly up-regulated in P. gingivalis LPS- and CpG-ODN-treated B cells from WT and TLR2 KO but not TLR4 KO mice. These results suggested that both TLR2 and TLR4 signaling are required for P. gingivalis LPS-induced, CpG-ODN-enhanced suppression of innate-like B cell apoptosis.
Collapse
Affiliation(s)
- Xiaoqian Yu
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
- Peking University School and Hospital of Stomatology, Department of Periodontology, Beijing, China
| | - Yuhua Wang
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
- Ninth People’s Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, Department of Prosthodontics, Shanghai Key laboratory, Shanghai, China
| | - Jiang Lin
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
- The Fourth Hospital of Harbin Medical University, Department of stomatology, Harbin, China
| | - Yang Hu
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
| | - Toshihisa Kawai
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
| | - Martin A. Taubman
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
| | - Xiaozhe Han
- The Forsyth Institute, Department of Immunology and Infectious Diseases, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
8
|
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
|
9
|
Altered circulating leukocytes and their chemokines in a clinical trial of therapeutic hypothermia for neonatal hypoxic ischemic encephalopathy*. Pediatr Crit Care Med 2013; 14:786-95. [PMID: 23897243 DOI: 10.1097/pcc.0b013e3182975cc9] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To determine systemic hypothermia's effect on circulating immune cells and their corresponding chemokines after hypoxic ischemic encephalopathy in neonates. DESIGN In our randomized, controlled, multicenter trial of systemic hypothermia in neonatal hypoxic ischemic encephalopathy, we measured total and leukocyte subset and serum chemokine levels over time in both hypothermia and normothermia groups, as primary outcomes for safety. SETTING Neonatal ICUs participating in a Neurological Disorders and Stroke sponsored clinical trial of therapeutic hypothermia. PATIENTS Sixty-five neonates with moderate to severe hypoxic ischemic encephalopathy within 6 hours after birth. INTERVENTIONS Patients were randomized to normothermia of 37°C or systemic hypothermia of 33°C for 48 hours. MEASUREMENTS AND MAIN RESULTS Complete and differential leukocyte counts and serum chemokines were measured every 12 hours for 72 hours. The hypothermia group had significantly lower median circulating total WBC and leukocyte subclasses than the normothermia group before rewarming, with a nadir at 36 hours. Only the absolute neutrophil count rebounded after rewarming in the hypothermia group. Chemokines, monocyte chemotactic protein-1 and interleukin-8, which mediate leukocyte chemotaxis as well as bone marrow suppression, were negatively correlated with their target leukocytes in the hypothermia group, suggesting active chemokine and leukocyte modulation by hypothermia. Relative leukopenia at 60-72 hours correlated with an adverse outcome in the hypothermia group. CONCLUSIONS Our data are consistent with chemokine-associated systemic immunosuppression with hypothermia treatment. In hypothermic neonates, persistence of lower leukocyte counts after rewarming is observed in infants with more severe CNS injury.
Collapse
|
10
|
Citronberg J, Bostick R, Ahearn T, Turgeon DK, Ruffin MT, Djuric Z, Sen A, Brenner DE, Zick SM. Effects of ginger supplementation on cell-cycle biomarkers in the normal-appearing colonic mucosa of patients at increased risk for colorectal cancer: results from a pilot, randomized, and controlled trial. Cancer Prev Res (Phila) 2013; 6:271-81. [PMID: 23303903 PMCID: PMC3618532 DOI: 10.1158/1940-6207.capr-12-0327] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To estimate the effects of ginger on apoptosis, proliferation, and differentiation in the normal-appearing colonic mucosa, we randomized 20 people at increased risk for colorectal cancer to 2.0 g of ginger or placebo daily for 28 days in a pilot trial. Overall expression and distributions of Bax, Bcl-2, p21, hTERT, and MIB-1 (Ki-67) in colorectal crypts in rectal mucosa biopsies were measured using automated immunohistochemistry and quantitative image analysis. Relative to placebo, Bax expression in the ginger group decreased 15.6% (P = 0.78) in the whole crypts, 6.6% (P = 0.95) in the upper 40% (differentiation zone) of crypts, and 21.7% (P = 0.67) in the lower 60% (proliferative zone) of crypts; however, there was a 19% increase (P = 0.14) in Bax expression in the upper 40% relative to the whole crypt. While p21 and Bcl-2 expression remained relatively unchanged, hTERT expression in the whole crypts decreased by 41.2% (P = 0.05); the estimated treatment effect on hTERT expression was larger in the upper 40% of crypts (-47.9%; P = 0.04). In the ginger group, MIB-1 expression decreased in the whole crypts, upper 40% of crypts, and lower 60% of crypts by 16.9% (P = 0.39), 46.8% (P = 0.39), and 15.3% (P = 0.41), respectively. These pilot study results suggest that ginger may reduce proliferation in the normal-appearing colorectal epithelium and increase apoptosis and differentiation relative to proliferation--especially in the differentiation zone of the crypts and support a larger study to further investigate these results.
Collapse
Affiliation(s)
| | - Roberd Bostick
- Department of Epidemiology, Emory University, Atlanta, GA
- Winship Cancer Institute, Emory University, Atlanta, GA
| | - Thomas Ahearn
- Department of Epidemiology, Emory University, Atlanta, GA
| | - D. Kim Turgeon
- Department of Internal Medicine, University of Michigan Medical School
| | - Mack T. Ruffin
- Department of Family Medicine, University of Michigan Medical School
| | - Zora Djuric
- Department of Family Medicine, University of Michigan Medical School
| | - Ananda Sen
- Department of Family Medicine, University of Michigan Medical School
| | - Dean E. Brenner
- Department of Internal Medicine, University of Michigan Medical School
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
- VA Medical Center, Ann Arbor, MI
| | - Suzanna M. Zick
- Department of Family Medicine, University of Michigan Medical School
| |
Collapse
|
11
|
White GE, Iqbal AJ, Greaves DR. CC chemokine receptors and chronic inflammation--therapeutic opportunities and pharmacological challenges. Pharmacol Rev 2013; 65:47-89. [PMID: 23300131 DOI: 10.1124/pr.111.005074] [Citation(s) in RCA: 217] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Chemokines are a family of low molecular weight proteins with an essential role in leukocyte trafficking during both homeostasis and inflammation. The CC class of chemokines consists of at least 28 members (CCL1-28) that signal through 10 known chemokine receptors (CCR1-10). CC chemokine receptors are expressed predominantly by T cells and monocyte-macrophages, cell types associated predominantly with chronic inflammation occurring over weeks or years. Chronic inflammatory diseases including rheumatoid arthritis, atherosclerosis, and metabolic syndrome are characterized by continued leukocyte infiltration into the inflammatory site, driven in large part by excessive chemokine production. Over years or decades, persistent inflammation may lead to loss of tissue architecture and function, causing severe disability or, in the case of atherosclerosis, fatal outcomes such as myocardial infarction or stroke. Despite the existence of several clinical strategies for targeting chronic inflammation, these diseases remain significant causes of morbidity and mortality globally, with a concomitant economic impact. Thus, the development of novel therapeutic agents for the treatment of chronic inflammatory disease continues to be a priority. In this review we introduce CC chemokine receptors as critical mediators of chronic inflammatory responses and explore their potential role as pharmacological targets. We discuss functions of individual CC chemokine receptors based on in vitro pharmacological data as well as transgenic animal studies. Focusing on three key forms of chronic inflammation--rheumatoid arthritis, atherosclerosis, and metabolic syndrome--we describe the pathologic function of CC chemokine receptors and their possible relevance as therapeutic targets.
Collapse
Affiliation(s)
- Gemma E White
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | | |
Collapse
|
12
|
Nam BY, Paeng J, Kim SH, Lee SH, Kim DH, Kang HY, Li JJ, Kwak SJ, Park JT, Yoo TH, Han SH, Kim DK, Kang SW. The MCP-1/CCR2 axis in podocytes is involved in apoptosis induced by diabetic conditions. Apoptosis 2011; 17:1-13. [DOI: 10.1007/s10495-011-0661-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
13
|
De Molfetta GA, Lucíola Zanette D, Alexandre Panepucci R, Dos Santos ARD, da Silva WA, Antonio Zago M. Role of NFKB2 on the early myeloid differentiation of CD34+ hematopoietic stem/progenitor cells. Differentiation 2010; 80:195-203. [PMID: 20708837 DOI: 10.1016/j.diff.2010.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 06/02/2010] [Accepted: 07/24/2010] [Indexed: 12/11/2022]
Abstract
To better understand the early events regulating lineage-specific hematopoietic differentiation, we analyzed the transcriptional profiles of CD34+ human hematopoietic stem and progenitor cells (HSPCs) subjected to differentiation stimulus. CD34+ cells were cultured for 12 and 40h in liquid cultures with supplemented media favoring myeloid or erythroid commitment. Serial analysis of gene expression (SAGE) was employed to generate four independent libraries. By analyzing the differentially expressed regulated transcripts between the un-stimulated and the stimulated CD34+ cells, we observed a set of genes that was initially up-regulated at 12h but were then down-regulated at 40h, exclusively after myeloid stimulus. Among those we found transcripts for NFKB2, RELB, IL1B, LTB, LTBR, TNFRSF4, TGFB1, and IKBKA. Also, the inhibitor NFKBIA (IKBA) was more expressed at 12h. All those transcripts code for signaling proteins of the nuclear factor kappa B pathway. NFKB2 is a subunit of the NF-κB transcription factor that with RELB mediates the non-canonical NF-κB pathway. Interference RNA (RNAi) against NFKB1, NFKB2 and control RNAi were transfected into bone marrow CD34+HSPC. The percentage and the size of the myeloid colonies derived from the CD34+ cells decreased after inhibition of NFKB2. Altogether, our results indicate that NFKB2 gene has a role in the early commitment of CD34+HSPC towards the myeloid lineage.
Collapse
Affiliation(s)
- Greice Andreotti De Molfetta
- Center for Cell Therapy, Blood Center of the University of Sao Paulo, Rua Tenente Catão Roxo 2501, 14051-140 Ribeirão Preto - SP, Brazil
| | | | | | | | | | | |
Collapse
|
14
|
Si Y, Tsou CL, Croft K, Charo IF. CCR2 mediates hematopoietic stem and progenitor cell trafficking to sites of inflammation in mice. J Clin Invest 2010; 120:1192-203. [PMID: 20234092 DOI: 10.1172/jci40310] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 01/20/2010] [Indexed: 12/12/2022] Open
Abstract
HSCs are BM-derived, self-renewing multipotent cells that develop into circulating blood cells. They have been implicated in the repair of inflamed parenchymal tissue, but the signals that regulate their trafficking to sites of inflammation are unknown. As monocytes are recruited to sites of inflammation via chemoattractants that activate CCR2 on their surface, we investigated whether HSCs are also recruited to sites of inflammation through CCR2. Initial analysis indicated that in mice, CCR2 was expressed on subsets of HSCs and hematopoietic progenitor cells (HPCs) and that freshly isolated primitive hematopoietic cells (Lin-c-Kit+ cells) responded to CCR2 ligands in vitro. In vivo analysis indicated that after instillation of thioglycollate to cause aseptic inflammation and after administration of acetaminophen to induce liver damage, endogenous HSCs/HPCs were actively recruited to the peritoneum and liver, respectively, in WT but not Ccr2-/- mice. HSCs/HPCs recovered from the peritoneum successfully engrafted into the BM of irradiated primary and secondary recipients, confirming their self renewal and multipotency. Importantly, administration of exogenous WT, but not Ccr2-/-, HSCs/HPCs accelerated resolution of acetaminophen-induced liver damage and triggered the expression of genes characteristic of the macrophage M2 or repair phenotype. These findings reveal what we believe to be a novel role for CCR2 in the homing of HSCs/HPCs to sites of inflammation and suggest new functions for chemokines in promoting tissue repair and regeneration.
Collapse
Affiliation(s)
- Yue Si
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, USA
| | | | | | | |
Collapse
|
15
|
Beltrán-Debón R, Alonso-Villaverde C, Aragonès G, Rodríguez-Medina I, Rull A, Micol V, Segura-Carretero A, Fernández-Gutiérrez A, Camps J, Joven J. The aqueous extract of Hibiscus sabdariffa calices modulates the production of monocyte chemoattractant protein-1 in humans. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2010; 17:186-191. [PMID: 19765963 DOI: 10.1016/j.phymed.2009.08.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 07/15/2009] [Accepted: 08/06/2009] [Indexed: 05/28/2023]
Abstract
Diet supplementation and/or modulation is an important strategy to significantly improve human health. The search of plants as additional sources of bioactive phenolic compounds is relevant in this context. The aqueous extract of Hibiscus sabdariffa is rich in anthocyanins and other phenolic compounds including hydroxycitric and chlorogenic acids. Using this extract we have shown an effective protection of cultured peripheral blood mononuclear cells from the cellular death induced by H(2)O(2) and a significant role in the production of inflammatory cytokines. In vitro, the extract promotes the production of IL-6 and IL-8 and decreases the concentration of MCP-1 in supernatants in a dose-dependent manner. In humans, the ingestion of an acute dose of the extract (10g) was well tolerated and decreased plasma MCP-1 concentrations significantly without further effects on other cytokines. This effect was not due to a concomitant increase in the antioxidant capacity of plasma. Instead, its mechanisms probably involve a direct inhibition of inflammatory and/or metabolic pathways responsible for MCP-1 production, and may be relevant in inflammatory and chronic conditions in which the role of MCP-1 is well established. If beneficial effects are confirmed in patients, Hibiscus sabdariffa could be considered a valuable traditional herbal medicine for the treatment of chronic inflammatory diseases with the advantage of being devoid of caloric value or potential alcohol toxicity.
Collapse
Affiliation(s)
- R Beltrán-Debón
- Centre de Recerca Biomèdica, Hospital Universitari de Sant Joan, IISPV, Universitat Rovira i Virgili, C/Sant Joan s/n, 43201 Reus, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Hematopoietic stem and progenitor cells: their mobilization and homing to bone marrow and peripheral tissue. Immunol Res 2009; 44:160-8. [PMID: 19340403 DOI: 10.1007/s12026-009-8109-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are a rare population of precursor cells that possess the capacity for self-renewal and multilineage differentiation. In the bone marrow (BM), HSPCs warrant blood cell homeostasis. In addition, they may also replenish tissue-resident myeloid cells and directly participate in innate immune responses once they home to peripheral tissues. In this review, we summarize recent data on the signaling molecules that modulate the mobilization of HSPCs from BM and their migration to peripheral tissues.
Collapse
|
17
|
Davoust N, Vuaillat C, Androdias G, Nataf S. From bone marrow to microglia: barriers and avenues. Trends Immunol 2008; 29:227-34. [PMID: 18396103 DOI: 10.1016/j.it.2008.01.010] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 01/30/2008] [Accepted: 01/30/2008] [Indexed: 12/25/2022]
Abstract
Microglia form a unique population of brain-resident macrophages. Although microglia have been involved in multiple disorders of the central nervous system (CNS), the issue of microglial renewal, under normal or pathological conditions, has been controversial. In mice, results from bone marrow chimera studies indicated that microglia are slowly but continuously replenished by bone marrow-derived cells. Moreover, such a microglial turnover was found to be greatly accelerated under multiple neurological conditions. However, recent works questioned the use of irradiation/reconstitution experiments to assess microglial turnover. Based on these different studies, we propose here a re-evaluation of microglia origin(s) in the inflamed CNS. We also discuss the therapeutic perspectives offered by the demonstration of an adult microglial lineage, from bone marrow to brain.
Collapse
Affiliation(s)
- Nathalie Davoust
- INSERM U851, IFR Biosciences, University of Lyon, 69007 Lyon, France
| | | | | | | |
Collapse
|
18
|
Opalek JM, Ali NA, Lobb JM, Hunter MG, Marsh CB. Alveolar macrophages lack CCR2 expression and do not migrate to CCL2. JOURNAL OF INFLAMMATION-LONDON 2007; 4:19. [PMID: 17888174 PMCID: PMC2099427 DOI: 10.1186/1476-9255-4-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 09/22/2007] [Indexed: 11/21/2022]
Abstract
Background The recruitment of mononuclear cells has important implications for tissue inflammation. Previous studies demonstrated enhanced CCR1 and CCR5 expression and decreased CCR2 expression during in vitro monocyte to macrophage differentiation. To date, no study examined the in vivo differences in chemokine receptor expression between human peripheral blood monocytes and alveolar macrophages. Methods We examined the expression of these receptors in human peripheral blood monocytes and alveolar macrophages using microarray analysis, reverse-transcriptase PCR, flow cytometry and migration analyses. Results In contrast to peripheral blood monocytes, alveolar macrophages did not express the CCL2 receptor, CCR2, and did not migrate toward CCL2. In contrast, monocytes and freshly isolated resident alveolar macrophages both migrated towards CCL3. However, up to 6-fold more monocytes migrated toward equivalent concentrations of CCL3 than did alveolar macrophages from the same donor. While peripheral blood monocytes expressed the CCL3 receptor, CCR1, alveolar macrophages expressed the alternate CCL3 receptor, CCR5. The addition of anti-CCR5 blocking antibodies completely abrogated CCL3-induced migration in alveolar macrophages, but did not affect the migration of peripheral blood monocytes. Conclusion These data support the specificity of CCL2 to selectively drive monocyte, but not alveolar macrophage recruitment to the lung and CCR5 as the primary macrophage receptor for CCL3.
Collapse
Affiliation(s)
- Judy M Opalek
- Department of Pathology, The Ohio State University, Columbus Ohio, USA
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, The Ohio State University and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - Naeem A Ali
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, The Ohio State University and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - Jennifer M Lobb
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, The Ohio State University and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - Melissa G Hunter
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, The Ohio State University and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| | - Clay B Marsh
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, The Ohio State University and Dorothy M. Davis Heart and Lung Research Institute, Columbus, Ohio, USA
| |
Collapse
|
19
|
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
|
20
|
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: 14] [Impact Index Per Article: 0.7] [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
|
21
|
Damodaran TV, Patel AG, Greenfield ST, Dressman HK, Lin SM, Abou-Donia MB. Gene expression profiles of the rat brain both immediately and 3 months following acute sarin exposure. Biochem Pharmacol 2005; 71:497-520. [PMID: 16376859 DOI: 10.1016/j.bcp.2005.10.051] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2005] [Revised: 10/07/2005] [Accepted: 10/10/2005] [Indexed: 10/25/2022]
Abstract
We have studied sarin-induced global gene expression patterns at an early time point (15 min; 0.5xLD50) and a later time point (3 months; 1xLD50) using Affymetrix: Rat Neurobiology U34 chips in male, Sprague-Dawley rats and have identified a total of 65 (early) and 38 (late) genes showing statistically significant alterations from control levels at 15 min and 3 months, respectively. At the early time point, those that are classified as ion channel, cytoskeletal and cell adhesion molecules, in addition to neuropeptides and their receptors predominated over all other groups. The other groups included: cholinergic signaling, calcium channel and binding proteins, transporters, chemokines, GABAnergic, glutamatergic, aspartate, catecholaminergic, nitric oxide synthase, purinergic, and serotonergic signaling molecules. At the late time point, genes that are classified as calcium channel and binding proteins, cytoskeletal and cell adhesion molecules and GABAnergic signaling molecules were most prominent. Seven molecules (Ania-9, Arrb-1, CX-3C, Gabab-1d, Nos-2a, Nrxn-1b, PDE2) were identified that showed altered persistent expression in both time points. Selected genes from each of these time points were further validated using semi quantitative RT-PCR approaches. Some of the genes that were identified in the present study have been shown to be involved in organophosphate-induced neurotoxicity by both other groups as well as ours. Principal component analysis (PCA) of the expression data from both time points was used for comparative analysis of the gene expression, which indicated that the changes in gene expression were a function of dose and time of euthanasia after the treatment. Our model also predicts that besides dose and duration of post-treatment period, age and possibly other factors may be playing important roles in the regulation of pathways, leading to the neurotoxicity.
Collapse
Affiliation(s)
- Tirupapuliyur V Damodaran
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
22
|
Guo Y, Graham-Evans B, Broxmeyer HE. Murine embryonic stem cells secrete cytokines/growth modulators that enhance cell survival/anti-apoptosis and stimulate colony formation of murine hematopoietic progenitor cells. Stem Cells 2005; 24:850-6. [PMID: 16339641 DOI: 10.1634/stemcells.2005-0457] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stromal cell-derived factor (SDF)-1/CXCL12, released by murine embryonic stem (ES) cells, enhances survival, chemotaxis, and hematopoietic differentiation of murine ES cells. Conditioned medium (CM) from murine ES cells growing in the presence of leukemia inhibitory factor (LIF) was generated while the ES cells were in an undifferentiated Oct-4 expressing state. ES cell-CM enhanced survival of normal murine bone marrow myeloid progenitors (CFU-GM) subjected to delayed growth factor addition in vitro and decreased apoptosis of murine bone marrow c-kit(+)lin- cells. ES CM contained interleukin (IL)-1alpha, IL-10, IL-11, macrophage-colony stimulating factor (CSF), oncostatin M, stem cell factor, vascular endothelial growth factor, as well as a number of chemokines and other proteins, some of which are known to enhance survival/anti-apoptosis of progenitors. Irradiation of ES cells enhanced release of some proteins and decreased release of others. IL-6, FGF-9, and TNF-alpha, not detected prior to irradiation was found after ES cells were irradiated. ES cell CM also stimulated CFU-GM colony formation. Thus, undifferentiated murine ES cells growing in the presence of LIF produce/release a number of biologically active interleukins, CSFs, chemokines, and other growth modulatory proteins, results which may be of physiological and/or practical significance.
Collapse
Affiliation(s)
- Ying Guo
- Department of Microbiology/Immunology, Walther Oncology Center, Indiana University School of Medicine, 950 West Walnut Street, R2-302, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
23
|
Pérez de Lema G, Maier H, Franz TJ, Escribese M, Chilla S, Segerer S, Camarasa N, Schmid H, Banas B, Kalaydjiev S, Busch DH, Pfeffer K, Mampaso F, Schlöndorff D, Luckow B. Chemokine receptor Ccr2 deficiency reduces renal disease and prolongs survival in MRL/lpr lupus-prone mice. J Am Soc Nephrol 2005; 16:3592-601. [PMID: 16267157 DOI: 10.1681/asn.2005040426] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
MRL/MpJ-Fas(lpr)/J (MRL/lpr) mice represent a well-established mouse model of human systemic lupus erythematosus. MRL/lpr mice homozygous for the spontaneous lymphoproliferation mutation (lpr) are characterized by systemic autoimmunity, massive lymphadenopathy associated with proliferation of aberrant T cells, splenomegaly, hypergammaglobulinemia, arthritis, and fatal immune complex-mediated glomerulonephritis. It was reported previously that steady-state mRNA levels for the chemokine (C-C motif) receptor 2 (Ccr2) continuously increase in kidneys of MRL/lpr mice. For examining the role of Ccr2 for development and progression of immune complex-mediated glomerulonephritis, Ccr2-deficient mice were generated and backcrossed onto the MRL/lpr genetic background. Ccr2-deficient MRL/lpr mice developed less lymphadenopathy, had less proteinuria, had reduced lesion scores, and had less infiltration by T cells and macrophages in the glomerular and tubulointerstitial compartment. Ccr2-deficient MRL/lpr mice survived significantly longer than MRL/lpr wild-type mice despite similar levels of circulating immunoglobulins and comparable immune complex depositions in the glomeruli of both groups. Anti-dsDNA antibody levels, however, were reduced in the absence of Ccr2. The frequency of CD8+ T cells in peripheral blood was significantly lower in Ccr2-deficient MRL/lpr mice. Thus Ccr2 deficiency influenced not only monocyte/macrophage and T cell infiltration in the kidney but also the systemic T cell response in MRL/lpr mice. These data suggest an important role for Ccr2 both in the general development of autoimmunity and in the renal involvement of the lupus-like disease. These results identify Ccr2 as an additional possible target for the treatment of lupus nephritis.
Collapse
|
24
|
Broxmeyer HE, Cooper S, Hangoc G, Kim CH. Stromal Cell-Derived Factor-1/CXCL12 Selectively Counteracts Inhibitory Effects of Myelosuppressive Chemokines on Hematopoietic Progenitor Cell Proliferation In Vitro. Stem Cells Dev 2005; 14:199-203. [PMID: 15910246 DOI: 10.1089/scd.2005.14.199] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A variety of cytokines and chemokines exert potent myelosuppressive effects that play a role in the maintenance of hematopoiesis, which, if unchecked, may result in pathological impairment of blood cell production. Processes that modulate these myelosuppressive effects are not well defined. Here we demonstrate that stromal cell-derived factor-1 (SDF-1/CXCL12), known for its ability to attract and to promote survival of hematopoietic progenitor cells (HPCs) and stem cells, blocks the effects of a broad range of myelosuppressive chemokines on proliferation of HPCs in vitro. The regulatory effects of SDF/CXCL12 on colony formation by mouse bone marrow granulocyte-macrophage (CFUGM), erythroid (BFU-E), and multipotential (CFU-GEMM) progenitor cells were assessed. These cells were stimulated to proliferate by combinations of growth factors, such that responses of immature HPCs could be assessed. SDF-1/CXCL12 potently blocked myelosuppressive responses induced by CCL2/MCP-1, CCL3/MIP-1alpha, CCL19/CKbeta-11, CCL25/TECK, CXCL4/PF4, CXCL8/IL-8, CXCL10/IP-10, and XCL1/Lymphotactin. However, SDF/CDL12 did not influence myelosuppression induced by tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, transforming growth factor (TGF)-beta or the iron-binding proteins H-ferritin or lactoferrin (LF). LF, previously shown to suppress release of growth factors, is shown here to also suppress proliferation of immature subsets of HPCs. HPCs from marrows of mice expressing an SDF-1/CXCL12 transgene were insensitive to inhibition by SDF/CXCL12-sensitive myelosuppressive chemokines, but not to SDF/CCL12-insensitive cytokines (TNF-alpha, IFN-gamma, TGF-beta, H-Ferritin, or LF). Thus, SDF-1/CXCL12 differentially and selectively regulates suppression of HPC proliferation by chemokines. These effects may counter myelosuppressive effects of certain chemokines in vivo, where proliferation of HPCs must be sustained.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | |
Collapse
|
25
|
Tao W, Hangoc G, Cooper S, Broxmeyer HE. SDF-1α/CXCL12 enhances retroviral-mediated gene transfer into immature subsets of human and murine hematopoietic progenitor cells. Gene Ther 2003; 11:61-9. [PMID: 14681698 DOI: 10.1038/sj.gt.3302127] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genetic modification of hematopoietic stem and progenitor cells has the potential to treat diseases affecting blood cells. Oncoretroviral vectors have been used for gene therapy; however, clinical success has been limited in part by low gene transfer efficiencies. We found that the presence of stromal-derived factor 1 (SDF-1alpha)/CXCL12 during retroviral transduction significantly enhanced, in a dose-dependent fashion, gene transfer into immature subsets of high proliferative human and murine hematopoietic progenitor cells. Murine mononuclear bone marrow cells and purified c-Kit(+)Lin(-) bone marrow cells were prestimulated and transduced with the bicistronic retroviral vector MIEG3 on Retronectin-coated surfaces in the presence and absence of SDF-1. SDF-1 enhanced gene transduction of murine bone marrow and c-Kit(+)Lin(-) cells by 35 and 29%, respectively. Moreover, SDF-1 enhanced transduction of progenitors in these populations by 121 and 107%, respectively. SDF-1 also enhanced transduction of human immature subsets of high proliferative progenitors present in either nonadherent mononuclear or CD34(+) umbilical cord blood cells. Transduction of hematopoietic progenitors was further increased by preloading Retronectin-coated plates with retrovirus using low-speed centrifugation followed by increasing cell-virus interactions through brief centrifugation during the transduction procedure. These results may be of clinical relevance.
Collapse
Affiliation(s)
- W Tao
- Department of Microbiology and Immunology, The Walther Oncology Center, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA
| | | | | | | |
Collapse
|
26
|
Rao AR, Quinones MP, Garavito E, Kalkonde Y, Jimenez F, Gibbons C, Perez J, Melby P, Kuziel W, Reddick RL, Ahuja SK, Ahuja SS. CC Chemokine Receptor 2 Expression in Donor Cells Serves an Essential Role in Graft-versus-Host-Disease. THE JOURNAL OF IMMUNOLOGY 2003; 171:4875-85. [PMID: 14568968 DOI: 10.4049/jimmunol.171.9.4875] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The complete repertoire of cellular and molecular determinants that influence graft-vs-host disease (GVHD) is not known. Using a well-established murine model of GVHD (B6-->bm12 mice), we sought to elucidate the role of the donor non-T cell compartment and molecular determinants therein in the pathogenesis of GVHD. In this model the acute GVHD-inducing effects of purified B6 wild-type (wt) CD4(+) T cells was inhibited by wt non-T cells in a dose-dependent manner. Paradoxically, unlike the chronic GVHD phenotype observed in bm12 mice transplanted with B6wt unfractionated splenocytes, bm12 recipients of B6ccr2-null unfractionated splenocytes developed acute GVHD and died of IFN-gamma-mediated bone marrow aplasia. This switch from chronic to acute GVHD was associated with increased target organ infiltration of activated CD4(+) T cells as well as enhanced expression of Th1/Th2 cytokines, chemokines, and the antiapoptotic factor bfl1. In vitro, ccr2(-/-) CD4(+) T cells in unfractionated splenocytes underwent significantly less activation-induced cell death than B6wt CD4(+) T cells, providing another potential mechanistic basis along with enhanced expression of bfl1 for the increased numbers of activated T cells in target organs of B6ccr2(-/-) splenocyte-->bm12 mice. Collectively, these findings have important clinical implications, as they implicate the donor non-T cell compartment as a critical regulator of GVHD and suggest that ccr2 expression in this cellular compartment may be an important molecular determinant of activation-induced cell death and GVHD pathogenesis.
Collapse
MESH Headings
- Acute Disease
- Anemia, Aplastic/genetics
- Anemia, Aplastic/immunology
- Anemia, Aplastic/mortality
- Anemia, Aplastic/pathology
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/transplantation
- Cell Separation
- Cells, Cultured
- Chemokines/biosynthesis
- Chronic Disease
- Cytokines/biosynthesis
- Down-Regulation/genetics
- Down-Regulation/immunology
- Graft Survival/genetics
- Graft Survival/immunology
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/mortality
- Graft vs Host Disease/pathology
- Interferon-gamma/biosynthesis
- Interferon-gamma/physiology
- Lymphocyte Activation/genetics
- Lymphocyte Transfusion/mortality
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, CCR2
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/physiology
- Spleen/cytology
- Spleen/metabolism
- Spleen/pathology
- Spleen/transplantation
Collapse
Affiliation(s)
- Arun R Rao
- South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio, TX 78284, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kim WJH, Chereshnev I, Gazdoiu M, Fallon JT, Rollins BJ, Taubman MB. MCP-1 deficiency is associated with reduced intimal hyperplasia after arterial injury. Biochem Biophys Res Commun 2003; 310:936-42. [PMID: 14550294 DOI: 10.1016/j.bbrc.2003.09.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocyte chemoattractant protein (MCP)-1 is abundant in smooth muscle cells (SMC) and macrophages of atherosclerotic plaques and in the injured arterial wall. MCP-1 and its receptor, CCR2, are important mediators of macrophage accumulation and atherosclerotic plaque progression. We have recently reported that CCR2(-/-) mice have a approximately 60% decrease in intimal hyperplasia and medial DNA synthesis in response to femoral arterial injury. We have now examined the response to femoral arterial injury in MCP-1(-/-) mice. MCP-1 deficiency was associated with a approximately 30% reduction in intimal hyperplasia at 4 weeks and was not associated with diminished medial DNA synthesis. Despite inducing tissue factor in SMC culture, MCP-1 deficiency was not associated with a decrease in neointimal tissue factor after injury. These data suggest that MCP-1 and CCR2 deficiencies have distinct effects on arterial injury. The effects of MCP-1 on intimal hyperplasia may be mediated largely through SMC migration.
Collapse
Affiliation(s)
- William J H Kim
- The Zena and Michael A Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | |
Collapse
|
28
|
Broxmeyer HE, Kohli L, Kim CH, Lee Y, Mantel C, Cooper S, Hangoc G, Shaheen M, Li X, Clapp DW. Stromal cell-derived factor-1/CXCL12 directly enhances survival/antiapoptosis of myeloid progenitor cells through CXCR4 and G(alpha)i proteins and enhances engraftment of competitive, repopulating stem cells. J Leukoc Biol 2003; 73:630-8. [PMID: 12714578 DOI: 10.1189/jlb.1002495] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Stromal cell-derived factor-1 (SDF-1/CXCL12) enhances survival of myeloid progenitor cells. The two main questions addressed by us were whether these effects on the progenitors were direct-acting and if SDF-1/CXCL12 enhanced engrafting capability of competitive, repopulating mouse stem cells subjected to short-term ex vivo culture with other growth factors. SDF-1/CXCL12 had survival-enhancing/antiapoptosis effects on human bone marrow (BM) and cord blood (CB) and mouse BM colony-forming units (CFU)-granulocyte macrophage, burst-forming units-erythroid, and CFU-granulocyte-erythroid-macrophage-megakaryocyte with similar dose responses. The survival effects were direct-acting, as assessed on colony formation by single isolated human BM and CB CD34(+++) cells. Effects were mediated through CXCR4 and G(alpha)i proteins. Moreover, SDF-1/CXCL12 greatly enhanced the engrafting capability of mouse long-term, marrow-competitive, repopulating stem cells cultured ex vivo with interleukin-6 and steel factor for 48 h. These results extend information on the survival effects mediated through the SDF-1/CXCL12-CXCR4 axis and may be of relevance for ex vivo expansion and gene-transduction procedures.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis 46202, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Broxmeyer HE, Cooper S, Kohli L, Hangoc G, Lee Y, Mantel C, Clapp DW, Kim CH. Transgenic expression of stromal cell-derived factor-1/CXC chemokine ligand 12 enhances myeloid progenitor cell survival/antiapoptosis in vitro in response to growth factor withdrawal and enhances myelopoiesis in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:421-9. [PMID: 12496427 DOI: 10.4049/jimmunol.170.1.421] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hemopoiesis is regulated in part by survival/apoptosis of hemopoietic stem/progenitor cells. Exogenously added stromal cell-derived factor-1 ((SDF-1)/CXC chemokine ligand (CXCL)12) enhances survival/antiapoptosis of myeloid progenitor cells in vitro. To further evaluate SDF-1/CXCL12 effects on progenitor cell survival, transgenic mice endogenously expressing SDF-1/CXCL12 under a Rous sarcoma virus promoter were produced. Myeloid progenitors (CFU-granulocyte-macrophage, burst-forming unit-erythroid, CFU-granulocyte-erythrocyte-megakaryocyte-monocyte) from transgenic mice were studied for in vitro survival in the context of delayed addition of growth factors. SDF-1-expressing transgenic myeloid progenitors were enhanced in survival and antiapoptosis compared with their wild-type littermate counterparts. Survival-enhancing effects were due to release of low levels of SDF-1/CXCL12 and mediated through CXCR4 and G(alpha)i proteins as determined by ELISA, an antagonist to CXCR4, Abs to CXCR4 and SDF-1, and pertussis toxin. Transgenic effects of low SDF-1/CXCR4 may be due to synergy of SDF-1/CXCL12 with other cytokines; low SDF-1/CXCL12 synergizes with low concentrations of other cytokines to enhance survival of normal mouse myeloid progenitors. Consistent with in vitro results, progenitors from SDF-1/CXCL12 transgenic mice displayed enhanced marrow and splenic myelopoiesis: greatly increased progenitor cell cycling and significant increases in progenitor cell numbers. These results substantiate survival effects of SDF-1/CXCL12, now extended to progenitors engineered to endogenously produce low levels of this cytokine, and demonstrate activity in vivo for SDF-1/CXCL12 in addition to cell trafficking.
Collapse
MESH Headings
- Adjuvants, Immunologic/antagonists & inhibitors
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/physiology
- Animals
- Apoptosis/genetics
- Apoptosis/immunology
- Bone Marrow Cells/cytology
- Bone Marrow Cells/immunology
- Cell Survival/genetics
- Cell Survival/immunology
- Chemokine CXCL12
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/immunology
- Colony-Forming Units Assay
- Cytokines/physiology
- Dose-Response Relationship, Immunologic
- Drug Synergism
- Gene Expression Regulation/immunology
- Growth Substances/metabolism
- Immune Sera/pharmacology
- Mice
- Mice, Inbred C3H
- Mice, Transgenic
- Myeloid Progenitor Cells/cytology
- Myeloid Progenitor Cells/immunology
- Myelopoiesis/genetics
- Myelopoiesis/immunology
- Pertussis Toxin/pharmacology
- Stromal Cells/physiology
- Transgenes/immunology
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Borzi RM, Mazzetti I, Magagnoli G, Paoletti S, Uguccioni M, Gatti R, Orlandini G, Cattini L, Facchini A. Growth-related oncogene alpha induction of apoptosis in osteoarthritis chondrocytes. ARTHRITIS AND RHEUMATISM 2002; 46:3201-11. [PMID: 12483724 DOI: 10.1002/art.10650] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To evaluate the apoptotic effect of the chemokine growth-related oncogene alpha (GROalpha), which we recently reported to be up-regulated in osteoarthritis (OA) chondrocytes. Chondrocyte apoptosis is considered to be a major determinant of cartilage damage in OA, a disease resulting from the aberrant production of inflammatory mediators (cytokines and chemokines) and effectors (matrix metalloproteinases and reactive oxygen and nitrogen species) by chondrocytes. METHODS We investigated the apoptotic effect of GROalpha on isolated human cells and on in vitro-cultured cartilage explants by conventional methods (morphology, detection of DNA fragmentation in situ and in solution, exposure of phosphatidylserine) and by analysis of "early" biochemical events (plasma membrane depolarization, activation of caspase 3, and phosphorylation of c-Jun N-terminal kinase/stress-activated protein kinase). RESULTS We clearly demonstrated that GROalpha was able to initiate a series of morphologic, biochemical, and molecular changes that led to chondrocyte apoptosis. Moreover, we found that additional signals delivered from the extracellular matrix (ECM) were essential in the control of chondrocyte susceptibility to GROalpha-induced apoptosis, since cell death was detected only when cells were stimulated after reestablishment of their proper interactions with the ECM, or in cartilage explant samples with reduced ECM, as indicated by decreased Safranin O staining. CONCLUSION GROalpha can induce apoptosis in articular chondrocytes, and the induction is dependent upon additional signals from the ECM. These findings are relevant to understanding the pathogenesis of OA, in view of the availability of the GROalpha chemokine in the joint space in the course of this rheumatic disease.
Collapse
|
31
|
Biswas SK, Sodhi A. Effect of monocyte chemoattractant protein-1 on murine bone marrow cells: proliferation, colony-forming ability and signal transduction pathway involved. Int J Immunopathol Pharmacol 2002; 15:183-194. [PMID: 12575918 DOI: 10.1177/039463200201500304] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) plays a crucial role in the migration and activation of leukocytes in both physiological and pathological contexts. In this paper, we report the in vitro effect of MCP-1 on myeloid haematopoiesis. MCP-1-treated murine nonadherent bone marrow cells (NABMCs) were assayed for in vitro proliferation and colony forming ability. It is observed that MCP-1 treatment in vitro caused an enhancement in the proliferation and colony forming ability of the murine NABMCs as compared to the untreated cells. This response was concentration-dependent and most effective at a dose of 100ng/ml MCP-1. In the presence of MCSF (200U/ml), GCSF (200U/ml), GMCSF (200U/ml) or IL-3 (200U/ml), the MCP-1-induced colony forming ability of the NABMCs was significantly augmented, indicating a synergistic effect of MCP-1 with these CSFs. However, irrespective of the CSFs used, MCP-1 stimulated the lineage-restricted differentiation of the murine BMCs into predominantly the granulocytic lineage. NABMCs cultured in medium alone formed minimal colonies. The probable signal transduction mechanism responsible for the MCP-1-induced NABMC proliferation/differentiation was also investigated. The results of the colony forming assay indicate that the protein kinase inhibitors, genistein (10&mgr;g/ml), chelenthryin chloride (10&mgr;M), wortmannin (200nM) and PD98059 (10&mgr;M) significantly blocked the in vitro colony forming ability of the MCP-1-treated NABMCs, while the phosphatase inhibitors, okadaic acid (10nM) and sodium orthovanadate (10&mgr;M) caused an increase in the BMC colony forming ability in response to MCP-1. These data suggests the involvement of the respective protein kinases and phosphatases in the above process. Correlating with this, the role of several signaling molecules likes Lyn, p42/44MAPK, PI3K and STAT5 has also been implicated in the signal cascade of murine NABMC proliferation/differentiation following MCP-1 treatment.
Collapse
Affiliation(s)
- S. K. Biswas
- School of Biotechnology, Banaras Hindu University, Varanasi, India
| | | |
Collapse
|
32
|
Basu S, Hodgson G, Katz M, Dunn AR. Evaluation of role of G-CSF in the production, survival, and release of neutrophils from bone marrow into circulation. Blood 2002; 100:854-61. [PMID: 12130495 DOI: 10.1182/blood.v100.3.854] [Citation(s) in RCA: 220] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In steady-state hematopoiesis, G-CSF (granulocyte-colony stimulating factor) regulates the level of neutrophils in the bone marrow and blood. In this study, we have exploited the availability of G-CSF-deficient mice to evaluate the role of G-CSF in steady-state granulopoiesis and the release of granulocytes from marrow into circulation. The thymidine analogue bromodeoxyuridine (BrdU) was used to label dividing bone marrow cells, allowing us to follow the release of granulocytes into circulation. Interestingly, the labeling index and the amount of BrdU incorporated by blast cells in bone marrow was greater in G-CSF-deficient mice than in wild-type mice. In blood, 2 different populations of BrdU-positive granulocytes, BrdU(bright) and BrdU(dim), could be detected. The kinetics of release of the BrdU(bright) granulocytes from bone marrow into blood was similar in wild-type and G-CSF-deficient mice; however, BrdU(dim) granulocytes peaked earlier in G-CSF-deficient mice. Our findings suggest that the mean transit time of granulocytes through the postmitotic pool is similar in G-CSF-deficient and control mice, although the transit time through the mitotic pool is reduced in G-CSF-deficient mice. Moreover, the reduced numbers of granulocytes that characterize G-CSF-deficient mice is primarily due to increased apoptosis in cells within the granulocytic lineage. Collectively, our data suggest that at steady state, G-CSF is critical for the survival of granulocytic cells; however, it is dispensable for trafficking of granulocytes from bone marrow into circulation.
Collapse
Affiliation(s)
- Sunanda Basu
- Molecular Biology Laboratory, Ludwig Institute for Cancer Research, Royal Melbourne Hospital, Victoria, Australia.
| | | | | | | |
Collapse
|
33
|
Fukuda S, Wu DW, Stark K, Pelus LM. Cloning and characterization of a proliferation-associated cytokine-inducible protein, CIP29. Biochem Biophys Res Commun 2002; 292:593-600. [PMID: 11922608 DOI: 10.1006/bbrc.2002.6680] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We identified a novel erythropoietin (Epo)-induced protein (CIP29) in lysates of human UT-7/Epo leukemia cells using two-dimensional gel analysis and cloned its full-length cDNA. CIP29 contains 210 amino acids with a predicted MW of 24 kDa, and has a N-terminal SAP DNA-binding motif. CIP29 expression was higher in cancer and fetal tissues than in normal adult tissues. CIP29 mRNA expression is cytokine regulated in hematopoietic cells, being up-regulated by Epo in UT7/Epo cells, and by thrombopoietin (Tpo), FLT3 ligand (FL) and stem cell factor (SCF) in primary human CD34(+) cells. Up-regulation of CIP29 in UT7/Epo cells by Epo was associated with cell cycle progression but not with antiapoptosis. Epo withdrawal reduced CIP29 expression concomitant with cell cycle arrest. Overexpression of CIP29-GFP in HEK293 cells enhances cell cycle progression. CIP29 appears to be a new cytokine regulated protein involved in normal and cancer cell proliferation.
Collapse
Affiliation(s)
- Seiji Fukuda
- Department of Microbiology and Immunology and Walther Oncology Center, Indiana University School of Medicine and Walther Cancer Institute, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
34
|
Roque M, Kim WJH, Gazdoin M, Malik A, Reis ED, Fallon JT, Badimon JJ, Charo IF, Taubman MB. CCR2 deficiency decreases intimal hyperplasia after arterial injury. Arterioscler Thromb Vasc Biol 2002; 22:554-9. [PMID: 11950690 DOI: 10.1161/hq0402.105720] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monocyte chemoattractant protein (MCP)-1 is upregulated in atherosclerotic plaques and in the media and intima of injured arteries. CC chemokine receptor 2 (CCR2) is the only known functional receptor for MCP-1. Mice deficient in MCP-1 or CCR2 have marked reductions in atherosclerosis. This study examines the effect of CCR2 deficiency in a murine model of femoral arterial injury. Four weeks after injury, arteries from CCR2(-/-) mice showed a 61.4% reduction (P<0.01) in intimal area and a 62% reduction (P<0.05) in intima/media ratio when compared with CCR2(+/+) littermates. The response of CCR2(+/-) mice was not significantly different from that of CCR2(+/+) mice. Five days after injury, the medial proliferation index, determined by bromodeoxyuridine incorporation, was decreased by 59.8% in CCR2(-/-) mice when compared with CCR2(+/+) littermates (P<0.05). Although leukocytes rapidly adhered to the injured arterial surface, there was no significant macrophage infiltration in the arterial wall of either CCR2(-/-) or CCR2(+/+) mice 5 and 28 days after injury. These results demonstrate that CCR2 plays an important role in mediating smooth muscle cell proliferation and intimal hyperplasia in a non-hyperlipidemic model of acute arterial injury. CCR2 may thus be an important target for inhibiting the response to acute arterial injury.
Collapse
Affiliation(s)
- Merce Roque
- Zena and Michael A. Wiener Cardiovascular Institute, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
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
|
36
|
Broxmeyer HE, Youn BS, Kim C, Hangoc G, Cooper S, Mantel C. Chemokine regulation of hematopoiesis and the involvement of pertussis toxin-sensitive G alpha i proteins. Ann N Y Acad Sci 2001; 938:117-27; discussion 127-8. [PMID: 11458498 DOI: 10.1111/j.1749-6632.2001.tb03580.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chemokines have been implicated in regulation of various aspects of hematopoiesis, including negative regulation of the proliferation of immature subsets of myeloid progenitor cells (MPCs), chemotaxis of MPCs, and survival enhancement of MPCs after delayed growth factor addition. Since chemokine receptors are seven-transmembrane-spanning G-protein-linked receptors and the chemotactic effect in vitro of the CXC chemokine SDF-1 is pertussis toxin (PT)-sensitive, implying the involvement of G alpha i proteins as mediators of SDF-1-induced chemotaxis, we evaluated the effects of PT on other chemokine actions influencing MPCs. While the in vitro survival-enhancing effects of SDF-1 on GM-CSF and steel factor-dependent mouse bone marrow granulocyte macrophage progenitors (CFU-GM) were pertussis toxin-sensitive, the suppressive effects of the CC chemokine MIP-1 alpha and the CXC chemokine IL-8 on colony formation by GM-CSF and steel factor-sensitive CFU-GM were insensitive to pertussis toxin. These results suggest that not all chemokine-mediated effects on MPCs are necessarily mediated through pertussis toxin-sensitive G alpha i proteins.
Collapse
Affiliation(s)
- H E Broxmeyer
- Departments of Microbiology and Immunology, and Medicine (Hematology/Oncology), the Walther Oncology Center, Indiana University School of Medicine, and the Walther Cancer Institute, Indianapolis, Indiana 46202, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Verbeek W, Wächter M, Lekstrom-Himes J, Koeffler HP. C/EBPepsilon -/- mice: increased rate of myeloid proliferation and apoptosis. Leukemia 2001; 15:103-11. [PMID: 11243377 DOI: 10.1038/sj.leu.2401995] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
CCAAT/enhancer binding protein epsilon (C/EBPepsilon) is essential for terminal granulocytic differentiation. Its expression begins at the transition between the proliferative and non-proliferative compartments of myelopoiesis. We studied the effect of targeted disruption of the C/EBPepsilon gene on murine myeloid proliferation and apoptosis. Bone marrow cellularity of C/EBPepsilon -/- and wild-type mice was 95% and 65%, respectively. The C/EBPepsilon -/- mice had an expansion in the number of their CFU-GM/femur. The number of myeloid committed progenitor cells in the peripheral blood and the spleen of these mice was also increased. Bromodeoxyuridine (BrDU) pulse labeling studies demonstrated that the fraction of actively proliferating cells was two-fold higher in the bone marrow of C/EBPepsilon -/- mice. However, the number of myeloid colonies arising from purified Sca-1+/lin- early hematopoietic progenitor cells and from bone marrow mononuclear cells grown in different cytokine combinations was not significantly different between wild-type and knock-out mice. Also, long-term marrow growth, and CFU were not different between the wild-type and C/EBPepsilon -/- mice. The sensitivity to induction of apoptosis in the committed progenitor cell compartment after either withdrawal of growth factor or brief exposure to etoposide was normal. However, Gr-1 antigen-positive C/EBPepsilon -/- granulocytic cells showed an increased rate of apoptosis in comparison to their wild-type counterparts. In summary, the myeloid compartment appears to be expanded in mice lacking C/EBPepsilon. However, this is not the consequence of an intrinsic myeloproliferation but due to an indirect, possibly cytokine-mediated stimulation of myelopoiesis in vivo. C/EBPepsilon may have a role in the inhibition of apoptosis in maturing granulocytic cells.
Collapse
Affiliation(s)
- W Verbeek
- Division of Hematology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | |
Collapse
|
38
|
Rosu-Myles M, Khandaker M, Wu DM, Keeney M, Foley SR, Howson-Jan K, Yee IC, Fellows F, Kelvin D, Bhatia M. Characterization of chemokine receptors expressed in primitive blood cells during human hematopoietic ontogeny. Stem Cells 2000; 18:374-81. [PMID: 11007922 DOI: 10.1634/stemcells.18-5-374] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Chemokines are capable of regulating a variety of fundamental processes of hematopoietic cells that include proliferation, differentiation, and migration. To evaluate potential chemokine signaling pathways important to the regulation of primitive human hematopoietic cells, we examined chemokine receptor expression of highly purified subpopulations of uncommitted human blood cells. CXCR1-, CXCR2-, CXCR4-, and CCR5-expressing cells were detected by flow cytometry among human blood subsets depleted of lineage-restricted cells (Lin(-)) derived from adult bone marrow, mobilized peripheral blood, cord blood (CB), and circulating fetal blood. Although these chemokine receptors could be detected on Lin(-) cells throughout human development, only CXCR4 could be detected in CD34(-)CD38(-)Lin(-) and CD34(+)CD38(-)Lin(-) subfractions enriched for stem cell function, suggesting that independent of ontogeny, CXCR4-mediated signals are critical to primitive hematopoiesis. Distinct to other stages of human hematopoietic development, primitive CB cells expressed higher levels of CXCR1, CXCR2, CCR5, and CXCR4 on both CD34(-)CD38(-)Lin(-) and CD34(+)CD38(-)Lin(-) subsets. Isolation of these fractions revealed expression of additional chemokine receptors CCR7, CCR8, and Bonzo (STRL133), whereas BOB (GPR15) could not be detected. Our study illustrates that rare uncommitted hematopoietic cells express chemokine receptors not previously associated with primitive human blood cells. Based on these results, we suggest that signaling pathways mediated by chemokine receptors identified here may play a fundamental role in hematopoietic stem cell regulation and provide alternative receptor targets for retroviral pseudotyping for genetic modification of repopulating cells.
Collapse
MESH Headings
- Adult
- Antigens, CD/genetics
- Fetal Blood/physiology
- Fetus
- Gene Expression Regulation, Developmental
- Hematopoiesis/physiology
- Hematopoietic Stem Cell Mobilization
- Hematopoietic Stem Cells/immunology
- Humans
- Infant, Newborn
- Receptors, CCR5/genetics
- Receptors, CXCR4/genetics
- Receptors, Chemokine/genetics
- Receptors, Interleukin/genetics
- Receptors, Interleukin-8A
- Receptors, Interleukin-8B
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- M Rosu-Myles
- The John P. Robarts Research Institute, Developmental Stem Cell Biology, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang S, Fukuda S, Lee Y, Hangoc G, Cooper S, Spolski R, Leonard WJ, Broxmeyer HE. Essential role of signal transducer and activator of transcription (Stat)5a but not Stat5b for Flt3-dependent signaling. J Exp Med 2000; 192:719-28. [PMID: 10974037 PMCID: PMC2193267 DOI: 10.1084/jem.192.5.719] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2000] [Accepted: 07/17/2000] [Indexed: 11/17/2022] Open
Abstract
The receptor tyrosine kinase Flt3 plays an important role in proliferation and survival of hematopoietic stem and progenitor cells. Although some post-receptor signaling events of Flt3 have been characterized, the involvement of the Janus kinase/signal transducer and activator of transcription (Jak/Stat) pathway in Flt3 signaling has not been thoroughly evaluated. To this aim, we examined whether Flt3 activates the Jak/Stat pathway in Baf3/Flt3 cells, a line stably expressing human Flt3 receptor. Stat5a, but not Stats 1-4, 5b, or 6, was potently activated by Flt3 ligand (FL) stimulation. Interestingly, FL did not activate any Jaks. Activation of Stat5a required the kinase activity of Flt3. A selective role for Stat5a in the proliferative response of primary hematopoietic progenitor cells to FL was documented, as FL did not act on progenitors from marrows of Stat5a(-/-) mice, but did stimulate/costimulate proliferation of these cells from Stat5a(+/+), Stat5b(-/-), and Stat5b(+/+) mice. Thus, Stat5a is essential for at least certain effects of FL. Moreover, our data confirm that Stat5a and Stat5b are not redundant, but rather are at least partially distinctive in their function.
Collapse
Affiliation(s)
- Shuli Zhang
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| | - Seiji Fukuda
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| | - Younghee Lee
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| | - Giao Hangoc
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| | - Scott Cooper
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| | - Rosanne Spolski
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892
| | - Warren J. Leonard
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, Bethesda, Maryland 20892
| | - Hal E. Broxmeyer
- Department of Microbiology/Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Walther Cancer Institute, Indianapolis, Indiana 46206
| |
Collapse
|
40
|
Abstract
In the armoury of the immune system developed to combat the various micro-organisms that could invade the host, the neutrophil forms the first line of defence against rapidly dividing bacteria and fungi. However, as humans age they become more susceptible to infection with these microbes and this has been ascribed to a decline in immune status, termed immune senescence. Here we summarize the literature specifically concerning the attenuation of neutrophil function with age and the possible mechanisms underlying their reduced response to infectious agents.
Collapse
Affiliation(s)
- S Butcher
- MRC Centre for Immune Regulation, Division of Immunity and Infection, Birmingham University Medical School, Birmingham, UK
| | | | | |
Collapse
|
41
|
Offermanns S. Mammalian G-protein function in vivo: new insights through altered gene expression. Rev Physiol Biochem Pharmacol 2000; 140:63-133. [PMID: 10857398 DOI: 10.1007/bfb0035551] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- S Offermanns
- Institut für Pharmakologie, Universitätsklinikum Benjamin Franklin, Freie Universität Berlin, Germany
| |
Collapse
|
42
|
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
|
43
|
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: 82] [Impact Index Per Article: 3.2] [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
|
44
|
Mollinedo F, Borregaard N, Boxer LA. Novel trends in neutrophil structure, function and development. IMMUNOLOGY TODAY 1999; 20:535-7. [PMID: 10636641 DOI: 10.1016/s0167-5699(99)01500-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- F Mollinedo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Spain.
| | | | | |
Collapse
|
45
|
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: 128] [Impact Index Per Article: 4.9] [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
|
46
|
Broxmeyer HE, Cooper S, Hangoc G, Gao JL, Murphy PM. Dominant myelopoietic effector functions mediated by chemokine receptor CCR1. J Exp Med 1999; 189:1987-92. [PMID: 10377195 PMCID: PMC2192966 DOI: 10.1084/jem.189.12.1987] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/1999] [Revised: 04/23/1999] [Indexed: 11/08/2022] Open
Abstract
Macrophage inflammatory protein (MIP)-1alpha, a CC chemokine, enhances proliferation of mature subsets of myeloid progenitor cells (MPCs), suppresses proliferation of immature MPCs, and mobilizes mature and immature MPCs to the blood. MIP-1alpha binds at least three chemokine receptors. To determine if CCR1 was dominantly mediating the above activities of MIP-1alpha, CCR1-deficient (-/-) mice, produced by targeted gene disruption, were used. MIP-1alpha enhanced colony formation of marrow granulocyte/macrophage colony-forming units (CFU-GM), responsive to stimulation by granulocyte/macrophage colony-stimulating factor (GM-CSF), and CFU-M, responsive to stimulation by M-CSF, from littermate control CCR1(+/+) but not CCR1(-/-) mice. Moreover, MIP-1alpha did not mobilize MPCs to the blood or synergize with G-CSF in this effect in CCR1(-/-) mice. However, CCR1(-/-) mice were increased in sensitivity to MPC mobilizing effects of G-CSF. Multi-growth factor-stimulated MPCs in CCR1(-/-) and CCR1(+/+) marrow were equally sensitive to inhibition by MIP-1alpha. These results implicate CCR1 as a dominant receptor for MIP-1alpha enhancement of proliferation of lineage-committed MPCs and for mobilization of MPCs to the blood. CCR1 is not a dominant receptor for MIP-1alpha suppression of MPC proliferation, but it does negatively impact G-CSF-induced MPC mobilization.
Collapse
Affiliation(s)
- H E Broxmeyer
- Department of Microbiology/Immunology, the Department of Medicine, and the Walther Oncology Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA.
| | | | | | | | | |
Collapse
|