1
|
Ariail E, Biggs B, O'Flanagan R, Schneck JP. IL-7 Immunotherapies: Current Applications and Engineering Opportunities. Immunol Invest 2025:1-19. [PMID: 39981682 DOI: 10.1080/08820139.2025.2464055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
BACKGROUND IL-7 is a cytokine that plays a critical role in the development and proliferation of many different immune cells. IL-7 is notably important for the proper development and activity of T cells and B cells. Additionally, the cytokine plays a role in the function of natural killer cells and dendritic cells. Because of this innate biological activity, IL-7 has gained traction as a potential immunotherapy for multiple applications. METHODS We conducted a comprehensive literature review to explore the physiological role of IL-7 and current applications harnessing the biology of IL-7 as a therapeutic. We also investigated the ways in which IL-7 is being engineered to enhance its therapeutic potential. RESULTS Notably, IL-7 has demonstrated efficacy in adoptive cell therapy models and as a vaccine adjuvant. The cytokine has also been used as a treatment for sepsis and other chronic infections. To further enhance its therapeutic efficacy, IL-7 has been engineered by fusing the cytokine to antibody fragments or other bioactive or targeting molecules. These engineered IL-7 therapeutics seek to improve the cytokine's pharmacokinetic and immunological properties and reduce off-target effects. CONCLUSION IL-7 immunotherapies largely remain at the preclinical stage, but there is growing interest in IL-7's many therapeutic applications and increasing opportunities to further engineer the molecule for future clinical translation.
Collapse
Affiliation(s)
- Emily Ariail
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin Biggs
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rowan O'Flanagan
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan P Schneck
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
2
|
IL-7: Comprehensive review. Cytokine 2022; 160:156049. [DOI: 10.1016/j.cyto.2022.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
|
3
|
Santos MLS, Coimbra RS, Sousa TN, Guimarães LFF, Gomes MS, Amaral LR, Pereira DB, Fontes CJF, Hawwari I, Franklin BS, Carvalho LH. The Interface Between Inflammatory Mediators and MicroRNAs in Plasmodium vivax Severe Thrombocytopenia. Front Cell Infect Microbiol 2021; 11:631333. [PMID: 33791239 PMCID: PMC8005714 DOI: 10.3389/fcimb.2021.631333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/17/2021] [Indexed: 11/27/2022] Open
Abstract
Severe thrombocytopenia can be a determinant factor in the morbidity of Plasmodium vivax, the most widespread human malaria parasite. Although immune mechanisms may drive P. vivax-induced severe thrombocytopenia (PvST), the current data on the cytokine landscape in PvST is scarce and often conflicting. Here, we hypothesized that the analysis of the bidirectional circuit of inflammatory mediators and their regulatory miRNAs would lead to a better understanding of the mechanisms underlying PvST. For that, we combined Luminex proteomics, NanoString miRNA quantification, and machine learning to evaluate an extensive array of plasma mediators in uncomplicated P. vivax patients with different degrees of thrombocytopenia. Unsupervised clustering analysis identified a set of PvST-linked inflammatory (CXCL10, CCL4, and IL-18) and regulatory (IL-10, IL-1Ra, HGF) mediators. Among the mediators associated with PvST, IL-6 and IL-8 were critical to discriminate P. vivax subgroups, while CCL2 and IFN-γ from healthy controls. Supervised machine learning spotlighted IL-10 in P. vivax-mediated thrombocytopenia and provided evidence for a potential signaling route involving IL-8 and HGF. Finally, we identified a set of miRNAs capable of modulating these signaling pathways. In conclusion, the results place IL-10 and IL-8/HGF in the center of PvST and propose investigating these signaling pathways across the spectrum of malaria infections.
Collapse
Affiliation(s)
| | - Roney S. Coimbra
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Tais N. Sousa
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | | | - Matheus S. Gomes
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Laurence R. Amaral
- Laboratório de Bioinformática e Análises Moleculares, Rede Multidisciplinar de Pesquisa, Ciência e Tecnologia, Universidade Federal de Uberlândia, Patos de Minas, Brazil
| | - Dhelio B. Pereira
- Dep. Pesquisa Clínica e Medicina Translacional, Centro de Pesquisas em Medicina Tropical, Porto Velho, Brazil
| | - Cor J. F. Fontes
- Departamento de Clínica Médica, Universidade Federal de Mato Grosso, Cuiabá, Brazil
| | - Ibrahim Hawwari
- Medical Faculty, Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Bernardo S. Franklin
- Medical Faculty, Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Luzia H. Carvalho
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| |
Collapse
|
4
|
Han F, Hu R, Su M, Yu Y, Yang H, Lai L. A human recombinant IL-7/HGFβ hybrid cytokine enhances antitumor immunity in mice. Am J Cancer Res 2017; 7:1714-1723. [PMID: 28861327 PMCID: PMC5574943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/05/2017] [Indexed: 06/07/2023] Open
Abstract
We purified a hybrid cytokine that contains interleukin-7 (IL-7) and the beta-chain of hepatocyte growth factor (HGFβ) from a unique long-term murine bone marrow culture system. We have cloned and expressed the human form of IL-7/HGFβ in which the IL-7 and HGFβ genes are connected by a flexible linker to produce a single-chain recombinant human IL-7/HGFβ protein (hrIL-7/HGFβ). To determine whether hrIL-7/HGFβ has antitumor activity, we injected this hybrid cytokine into melanoma and colon cancer animal models, and then assessed the local tumor growth and tumor metastasis. We show here that in vivo administration of hrIL-7/HGFβ significantly inhibited the growth and metastasis of malignant melanoma and colon cancer in mice. The antitumor activity was involved in a marked increase in the number of tumor-infiltrating CD4+ and CD8+ T cells and activated dendritic cells. The immunological mechanism by which hrIL-7/HGFβ inhibits tumor growth was confirmed by its inability to inhibit tumor growth in vitro and in immunodeficient mice. Furthermore, immune cells from hrIL-7/HGFβ-treated cancer-bearing mice can be adoptively transferred into naïve mice to resist same tumor cell challenge. Therefore, hrIL-7/HGFβ has potential applications in the treatment of cancer patients.
Collapse
Affiliation(s)
- Feng Han
- The Affiliated Hospital of Guizhou Medical University, University of ConnecticutStorrs, CT, USA
| | - Rong Hu
- Department of Allied Health Sciences, University of ConnecticutStorrs, CT, USA
| | - Min Su
- Department of Allied Health Sciences, University of ConnecticutStorrs, CT, USA
| | - Yanni Yu
- The Affiliated Hospital of Guizhou Medical University, University of ConnecticutStorrs, CT, USA
| | - Hua Yang
- The Affiliated Hospital of Guizhou Medical University, University of ConnecticutStorrs, CT, USA
| | - Laijun Lai
- Department of Allied Health Sciences, University of ConnecticutStorrs, CT, USA
- University of Connecticut Stem Cell Institute, University of ConnecticutStorrs, CT, USA
| |
Collapse
|
5
|
Interleukin-7 and Immunosenescence. J Immunol Res 2017; 2017:4807853. [PMID: 28484723 PMCID: PMC5397725 DOI: 10.1155/2017/4807853] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/01/2017] [Accepted: 02/19/2017] [Indexed: 12/21/2022] Open
Abstract
The age of an individual is an important, independent risk factor for many of the most common diseases afflicting modern societies. Interleukin-7 (IL-7) plays a central, critical role in the homeostasis of the immune system. Recent studies support a critical role for IL-7 in the maintenance of a vigorous healthspan. We describe the role of IL-7 and its receptor in immunosenescence, the aging of the immune system. An understanding of the role that IL-7 plays in aging may permit parsimonious preventative or therapeutic solutions for diverse conditions. Perhaps IL-7 might be used to "tune" the immune system to optimize human healthspan and longevity.
Collapse
|
6
|
Song Y, Liu Y, Hu R, Su M, Rood D, Lai L. In Vivo Antitumor Activity of a Recombinant IL7/IL15 Hybrid Cytokine in Mice. Mol Cancer Ther 2016; 15:2413-2421. [PMID: 27474151 DOI: 10.1158/1535-7163.mct-16-0111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/15/2016] [Indexed: 11/16/2022]
Abstract
Both IL7 and IL15 have become important candidate immunomodulators for cancer treatment. However, IL7 or IL15 used alone suffers from shortcomings, such as short serum half-life and limited antitumor effect. We have cloned and expressed a recombinant (r) IL7/IL15 fusion protein in which IL7 and IL15 are linked by a flexible linker. We then compared the antitumor effect of rIL7/IL15 with the individual factors rIL7 and/or rIL15. We show here that rIL7/IL15 has a higher antitumor activity than the combination of the individual factors in both murine B16F10 melanoma and CT-26 colon cancer models. This was associated with a significant increase in tumor infiltration of T cells, DCs, and NK cells and a decrease in regulatory T cells (Tregs). In addition, rIL7/IL15-treated DCs had higher expression of costimulatory molecules CD80 and CD86. The higher antitumor activity of rIL7/IL15 is likely due to its longer in vivo half-life and different effects on immune cells. Our results suggest that rIL7/IL15 may offer a new tool to enhance antitumor immunity and treat cancer. Mol Cancer Ther; 15(10); 2413-21. ©2016 AACR.
Collapse
Affiliation(s)
- Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. Medical College, Three Gorges University, Yichang, China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
7
|
Hu R, Liu Y, Song Y, Su M, Lu X, Rood D, Lai L. Recombinant IL-7/HGFβ hybrid cytokine separates acute graft-versus-host-disease from graft-versus-tumour activity by altering donor T cell trafficking. Br J Haematol 2016; 175:505-516. [PMID: 27447780 DOI: 10.1111/bjh.14268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/07/2016] [Indexed: 11/29/2022]
Abstract
Given that donor T cells from a transplant contribute both the desired graft-versus-tumour (GVT) effect and detrimental graft-versus-host disease (GVHD), strategies to separate GVHD and GVT activity are a major clinical goal. We have previously demonstrated that in vivo administration of a recombinant (r)IL-7/HGFβ hybrid cytokine, consisting of interleukin-7 (IL-7, IL7) and the β-chain of hepatocyte growth factor (HGFβ), significantly inhibits the growth of cancer cells in murine tumour models. The antit-umour effect of rIL-7/HGFβ is related to a marked infiltration T cells in the tumour tissues. We have also shown that GVHD was not induced in rIL-7/HGFβ-treated T cell-depleted allogeneic haematopoietic stem cell transplantation (HSCT) recipients. We show here that, in T cell-replete allogeneic HSCT murine models, rIL-7/HGFβ attenuated acute GVHD (aGVHD), while promoting GVT activity. This was related to an alteration of donor T cell trafficking, with an increased infiltration of donor T cells into tumour tissues and the lympho-haematopoietic system but decreased number of the T cells in the GVHD target organs. Therefore, rIL-7/HGFβ may offer a new tool to alleviate aGVHD while prompting GVT, and to study the molecular regulation of T cell trafficking.
Collapse
Affiliation(s)
- Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA.,Guizhou Medical University, Guizhou, China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Xiuling Lu
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA. .,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
8
|
Ilangumaran S, Villalobos-Hernandez A, Bobbala D, Ramanathan S. The hepatocyte growth factor (HGF)–MET receptor tyrosine kinase signaling pathway: Diverse roles in modulating immune cell functions. Cytokine 2016; 82:125-39. [PMID: 26822708 DOI: 10.1016/j.cyto.2015.12.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/14/2022]
|
9
|
Jung WS, Han SM, Kim SM, Kim ME, Lee JS, Seo KW, Youn HY, Lee HW. Stimulatory effect of HGF-overexpressing adipose tissue-derived mesenchymal stem cells on thymus regeneration in a rat thymus involution model. Cell Biol Int 2014; 38:1106-17. [DOI: 10.1002/cbin.10306] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 04/14/2014] [Indexed: 01/07/2023]
Affiliation(s)
- Woo-Sung Jung
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-742 Republic of Korea
| | - Sei-Myoung Han
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-742 Republic of Korea
| | - Sung-Min Kim
- Division of Magnetic Resonance Research; Korea Basic Science Institute; Ochang Chungbuk Republic of Korea
| | - Mi-Eun Kim
- Department of Biology, College of Natural Sciences; Chosun University; Gwangju Republic of Korea
| | - Jun-Sik Lee
- Department of Biology, College of Natural Sciences; Chosun University; Gwangju Republic of Korea
| | - Kyoung-Won Seo
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Chungnam National University; 99 Daehakro Yuseoung gu Daejon 305-764 Republic of Korea
| | - Hwa-Young Youn
- Department of Veterinary Internal Medicine, College of Veterinary Medicine; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-742 Republic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-742 Republic of Korea
| | - Hee-Woo Lee
- Research Institute for Veterinary Science, College of Veterinary Medicine; Seoul National University; 1 Gwanak-ro Gwanak-gu Seoul 151-742 Republic of Korea
| |
Collapse
|
10
|
Lai L, Zhang M, Song Y, Rood D. Recombinant IL-7/HGFβ hybrid cytokine enhances T cell recovery in mice following allogeneic bone marrow transplantation. PLoS One 2013; 8:e82998. [PMID: 24349415 PMCID: PMC3861470 DOI: 10.1371/journal.pone.0082998] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 11/08/2013] [Indexed: 11/23/2022] Open
Abstract
T cell immunodeficiency is a major complication of bone marrow (BM) transplantation (BMT). Therefore, approaches to enhance T cell reconstitution after BMT are required. We have purified a hybrid cytokine, consisting of IL-7 and the β-chain of hepatocyte growth factor (HGFβ) (IL-7/HGFβ), from a unique long-term BM culture system. We have cloned and expressed the IL-7/HGFβ gene in which the IL-7 and HGFβ genes are connected by a flexible linker to generate rIL-7/HGFβ protein. Here, we show that rIL-7/HGFβ treatment enhances thymopoiesis after allogeneic BMT. Although rIL-7 treatment also enhances the number of thymocytes, rIL-7/HGFβ hybrid cytokine was more effective than was rIL-7 and the mechanisms by which rIL-7 and rIL-7/HGFβ increase the numbers of thymocytes are different. rIL-7 enhances the survival of double negative (DN), CD4 and CD8 single positive (SP) thymocytes. In contrast, rIL-7/HGFβ enhances the proliferation of the DN, SP thymocytes, as well as the survival of CD4 and CD8 double positive (DP) thymocytes. rIL-7/HGFβ treatment also increases the numbers of early thymocyte progenitors (ETPs) and thymic epithelial cells (TECs). The enhanced thymic reconstitution in the rIL-7/HGFβ-treated allogeneic BMT recipients results in increased number and functional activities of peripheral T cells. Graft-versus-host-disease (GVHD) is not induced in the rIL-7/HGFβ-treated BMT mice. Therefore, rIL-7/HGFβ may offer a new tool for the prevention and/or treatment of T cell immunodeficiency following BMT.
Collapse
Affiliation(s)
- Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, United States of America
- University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| | - Mingfeng Zhang
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, United States of America
| | - Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, United States of America
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut, United States of America
| |
Collapse
|
11
|
Lai L, Zhang M, Goldschneider I. Recombinant IL-7/HGFβ efficiently induces transplantable murine hematopoietic stem cells. J Clin Invest 2012; 122:3552-62. [PMID: 22996694 DOI: 10.1172/jci46055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/26/2012] [Indexed: 01/29/2023] Open
Abstract
Difficulty obtaining sufficient hematopoietic stem cells (HSCs) directly from the donor has limited the clinical use of HSC transplantation. Numerous attempts to stimulate the ex vivo growth of purified HSCs with cytokines and growth factors generally have induced only modest increases in HSC numbers while decreasing their in vivo reconstituting ability. We previously developed a recombinant single-chain form of a naturally occurring murine hybrid cytokine of IL-7 and the β chain of hepatocyte growth factor (rIL-7/HGFβ) that stimulates the in vitro proliferation and/or differentiation of common lymphoid progenitors, pre-pro-B cells, and hematopoietic progenitor cells (day 12 spleen colony-forming units) in cultures of mouse BM. Here we used the rIL-7/HGFβ in culture to induce large numbers of HSCs from multiple cell sources, including unseparated BM cells, purified HSCs, CD45- BM cells, and embryonic stem cells. In each instance, most of the HSCs were in the G0 phase of the cell cycle and exhibited reduced oxidative stress, decreased apoptosis, and increased CXCR4 expression. Furthermore, when injected i.v., these HSCs migrated to BM, self-replicated, provided radioprotection, and established long-term hematopoietic reconstitution. These properties were amplified by injection of rIL-7/HGFβ directly into the BM cavity but not by treatment with rIL-7, rHGF, and/or rHGFβ.
Collapse
Affiliation(s)
- Laijun Lai
- Department of Immunology, University of Connecticut, Stem Cell Institute, School of Medicine, Health Center, Storrs, CT 06269, USA.
| | | | | |
Collapse
|
12
|
Mouse embryonic stem cell-derived thymic epithelial cell progenitors enhance T-cell reconstitution after allogeneic bone marrow transplantation. Blood 2011; 118:3410-8. [DOI: 10.1182/blood-2011-03-340794] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract
We have reported that mouse embryonic stem cells (mESCs) can be selectively induced in vitro to differentiate into thymic epithelial cell progenitors (TEPs). When placed in vivo, these mESC-derived TEPs differentiate into cortical and medullary thymic epithelial cells, reconstitute the normal thymic architecture, and enhance thymocyte regeneration after syngeneic BM transplantation (BMT). Here, we show that transplantation of mESC-derived TEPs results in the efficient establishment of thymocyte chimerism and subsequent generation of naive T cells in both young and old recipients of allo-geneic BM transplant. GVHD was not induced, whereas graft-versus-tumor activity was significantly enhanced. Importantly, the reconstituted immune system was tolerant to host, mESC, and BM transplant donor antigens. Therefore, ESC-derived TEPs may offer a new approach for the rapid and durable correction of T-cell immune deficiency after BMT, and the induction of tolerance to ESC-derived tissue and organ transplants. In addition, ESC-derived TEPs may also have use as a means to reverse age-dependent thymic involution, thereby enhancing immune function and decreasing infection rates in the elderly.
Collapse
|
13
|
A Human Recombinant IL-7/HGFα Hybrid Cytokine Enhances T-Cell Reconstitution in Mice After Syngeneic Bone Marrow Transplantation. Transplantation 2011; 92:516-22. [DOI: 10.1097/tp.0b013e318228d766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Jin J, Goldschneider I, Lai L. In Vivo Administration of the Recombinant IL-7/Hepatocyte Growth Factor β Hybrid Cytokine Efficiently Restores Thymopoiesis and Naive T Cell Generation in Lethally Irradiated Mice after Syngeneic Bone Marrow Transplantation. THE JOURNAL OF IMMUNOLOGY 2011; 186:1915-22. [DOI: 10.4049/jimmunol.1001238] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Olaharski AJ, Bitter H, Gonzaludo N, Kondru R, Goldstein DM, Zabka TS, Lin H, Singer T, Kolaja K. Modeling bone marrow toxicity using kinase structural motifs and the inhibition profiles of small molecular kinase inhibitors. Toxicol Sci 2010; 118:266-75. [PMID: 20810542 DOI: 10.1093/toxsci/kfq258] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The cellular function of kinases combined with the difficulty of designing selective small molecule kinase inhibitors (SMKIs) poses a challenge for drug development. The late-stage attrition of SMKIs could be lessened by integrating safety information of kinases into the lead optimization stage of drug development. Herein, a mathematical model to predict bone marrow toxicity (BMT) is presented which enables the rational design of SMKIs away from this safety liability. A specific example highlights how this model identifies critical structural modifications to avoid BMT. The model was built using a novel algorithm, which selects 19 representative kinases from a panel of 277 based upon their ATP-binding pocket sequences and ability to predict BMT in vivo for 48 SMKIs. A support vector machine classifier was trained on the selected kinases and accurately predicts BMT with 74% accuracy. The model provides an efficient method for understanding SMKI-induced in vivo BMT earlier in drug discovery.
Collapse
|
16
|
Lai L, Jin J. Generation of thymic epithelial cell progenitors by mouse embryonic stem cells. Stem Cells 2010; 27:3012-20. [PMID: 19824081 DOI: 10.1002/stem.238] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Thymopoiesisis regulated by the thymic microenvironment, of which epithelial cells are the major components. Both cortical and medullary thymic epithelial cells (TECs) have been shown to arise from a common progenitor cell. Here we show for the first time that mouse embryonic stem cells (mESCs) can be selectively induced in vitro to differentiate into cells that have the phenotype of thymic epithelial progenitors (TEPs). When placed in vivo, these mESC-derived TEPs self-renew, develop into TECs, and reconstitute the normal thymic architecture. Functionally, these ESC-derived TEPs enhanced thymocyte regeneration after bone marrow transplantation and increased the number of functional naive splenic T cells. In addition to providing a model to study the molecular events underlying thymic epithelial cell development, the ability to selectively induce the development of TEPs in vitro from mESCs has important implications regarding the prevention and/or treatment of primary and secondary T-cell immunodeficiencies.
Collapse
Affiliation(s)
- Laijun Lai
- Department of Immunology and, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | |
Collapse
|
17
|
Carrio R, Altman NH, Lopez DM. Downregulation of interleukin-7 and hepatocyte growth factor in the thymic microenvironment is associated with thymus involution in tumor-bearing mice. Cancer Immunol Immunother 2009; 58:2059-72. [PMID: 19421751 PMCID: PMC11030654 DOI: 10.1007/s00262-009-0714-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 04/17/2009] [Indexed: 12/18/2022]
Abstract
During mammary tumorigenesis, there is a profound thymic involution associated with severe depletion of the most abundant subset of thymocytes, CD4(+)CD8(+) immature cells, and an early arrest in at least two steps of T cell differentiation. Thymic atrophy that is normally related with aging has been observed in other model systems, including graft-vs-host disease (GVHD) and tumor development. However, the mechanisms involved in this phenomenon remain to be elucidated. Vascular endothelial growth factor (VEGF) has been associated with thymic involution, when expressed at high levels systemically. In thymuses of D1-DMBA-3 tumor-bearing mice, this growth factor is diminished relative to the level of normal thymuses. Interestingly, the expression of hepatocyte growth factor (HGF), which has been associated with proliferation, cell survival, angiogenesis and B-cell differentiation, is profoundly down-regulated in thymuses of tumor bearers. In parallel, IL-7 and IL-15 mRNA, crucial cytokines involved in thymocytes development and cellular homeostasis, respectively, are also down-regulated in the thymuses of tumor hosts as compared to those of normal mice. Injection of HGF into mice implanted with mammary tumors resulted in normalization of thymic volume and levels of VEGF, IL-7 and IL-15. While, injections of IL-7 partially restored the thymic involution observed in the thymuses of tumor-bearing mice, injection of IL-15 did not have any significant effects. Our data suggest that the downregulation of HGF and IL-7 may play an important role in the thymic involution observed in tumor-bearing hosts.
Collapse
Affiliation(s)
- Roberto Carrio
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, FL 33101 USA
| | - Norman H. Altman
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| | - Diana M. Lopez
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, FL 33101 USA
| |
Collapse
|
18
|
Sugiura K, Taketani S, Yoshimura T, Nishino T, Nishino N, Fujisawa JI, Hisha H, Inaba T, Ikehara S. Effect of hepatocyte growth factor on long term hematopoiesis of human progenitor cells in transgenic-sever combined immunodeficiency mice. Cytokine 2007; 37:218-26. [PMID: 17512212 DOI: 10.1016/j.cyto.2007.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Revised: 03/01/2007] [Accepted: 04/02/2007] [Indexed: 11/18/2022]
Abstract
Hepatocyte growth factor (HGF), which was originally isolated as a liver generating factor, enhances hematopoiesis. To study the effect of HGF on hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs), we generated severe combined immunodeficiency (SCID) mice producing human (h) HGF and/or stem cell factor (SCF) by transferring the relevant genes to fertilized eggs, and then transplanted hematopoietic progenitors from human cord blood into the transgenic (Tg) SCID mice. Six months after transplantation, a significantly larger number of human cells were found in the Tg SCID mice than in non-Tg controls. Characteristically, the recipient SCID mice producing h HGF (HGF-SCID) had a significantly increased number of h CD41+ cells, whereas the SCF-SCID recipients had more CD11b+ cells. Significantly large numbers of CD34+ progenitors were found in the SCID mice transferred with both h HGF and h SCF genes (HGF/SCF-SCID) when compared with HGF-SCID or SCF-SCID mice. These results imply that HGF supports the differentiation of progenitors in megakaryocyte lineage, whereas SCF supports that in myeloid lineage. The results also imply that HGF acts on HSCs/HPCs as a synergistic proliferative factor combined with SCF. We have demonstrated the advantage of the human cytokine-producing animal in the maintenance of human HSCs.
Collapse
Affiliation(s)
- Kikuya Sugiura
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai City, Osaka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|