51
|
Insights into the HIV Latency and the Role of Cytokines. Pathogens 2019; 8:pathogens8030137. [PMID: 31487807 PMCID: PMC6789648 DOI: 10.3390/pathogens8030137] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 09/01/2019] [Indexed: 12/23/2022] Open
Abstract
Human immunodeficiency virus-1 (HIV-1) has the ability to infect latently at the level of individual CD4+ cells. Latent HIV-1 proviruses are transcriptionally silent and immunologically inert, but are still capable of reactivating productive lytic infection following cellular activation. These latent viruses are the main obstacle in the eradication of HIV-1, because current HIV-1 treatment regimens are ineffective against them. Normal immunological response against an antigen activates CD4+ naïve T cells. The activated CD4+ naïve T cells undergo cell cycle, resulting in further transformation and profound proliferation to form effector CD4+ T-cells. Notably, in HIV-1 infected individuals, some of the effector CD4+ T cells get infected with HIV-1. Upon fulfillment of their effector functions, almost all activated CD4+ T cells are committed to apoptosis or programmed cell death, but a miniscule fraction revert to quiescence and become resting memory CD4+ T cells to mediate a rapid immunological response against the same antigen in the future. However, due to the quiescent nature of the resting memory T cells, the integrated HIV-1 becomes transcriptionally silent and acquires a latent phenotype. Following re-exposure to the same antigen, memory cells and integrated HIV-1 are stimulated. The reactivated latent HIV provirus subsequently proceeds through its life cycle and eventually leads to the production of new viral progeny. Recently, many strategies against HIV-1 latency have been developed and some of them have even matured to the clinical level, but none can yet effectively eliminate the latent HIV reservoir, which remains a barrier to HIV-1 cure. Therefore, alternative strategies to eradicate latent HIV need to be considered. This review provides vital knowledge on HIV latency and on strategies to supplement highly active anti-retroviral therapy (HAART) with cytokine-mediated therapeutics for dislodging the latent HIV reservoirs in order to open up new avenues for curing HIV.
Collapse
|
52
|
Arina A, Beckett M, Fernandez C, Zheng W, Pitroda S, Chmura SJ, Luke JJ, Forde M, Hou Y, Burnette B, Mauceri H, Lowy I, Sims T, Khodarev N, Fu YX, Weichselbaum RR. Tumor-reprogrammed resident T cells resist radiation to control tumors. Nat Commun 2019; 10:3959. [PMID: 31477729 PMCID: PMC6718618 DOI: 10.1038/s41467-019-11906-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Successful combinations of radiotherapy and immunotherapy depend on the presence of live T cells within the tumor; however, radiotherapy is believed to damage T cells. Here, based on longitudinal in vivo imaging and functional analysis, we report that a large proportion of T cells survive clinically relevant doses of radiation and show increased motility, and higher production of interferon gamma, compared with T cells from unirradiated tumors. Irradiated intratumoral T cells can mediate tumor control without newly-infiltrating T cells. Transcriptomic analysis suggests T cell reprogramming in the tumor microenvironment and similarities with tissue-resident memory T cells, which are more radio-resistant than circulating/lymphoid tissue T cells. TGFβ is a key upstream regulator of T cell reprogramming and contributes to intratumoral Tcell radio-resistance. These findings have implications for the design of radio-immunotherapy trials in that local irradiation is not inherently immunosuppressive, and irradiation of multiple tumors might optimize systemic effects of radiotherapy.
Collapse
Affiliation(s)
- Ainhoa Arina
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA.
| | - Michael Beckett
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Christian Fernandez
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenxin Zheng
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Steven J Chmura
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Jason J Luke
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Martin Forde
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Yuzhu Hou
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Byron Burnette
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Helena Mauceri
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Israel Lowy
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Tasha Sims
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Nikolai Khodarev
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
53
|
Roles of Myosin-Mediated Membrane Trafficking in TGF-β Signaling. Int J Mol Sci 2019; 20:ijms20163913. [PMID: 31408934 PMCID: PMC6719161 DOI: 10.3390/ijms20163913] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022] Open
Abstract
Recent findings have revealed the role of membrane traffic in the signaling of transforming growth factor-β (TGF-β). These findings originate from the pivotal function of TGF-β in development, cell proliferation, tumor metastasis, and many other processes essential in malignancy. Actin and unconventional myosin have crucial roles in subcellular trafficking of receptors; research has also revealed a growing number of unconventional myosins that have crucial roles in TGF-β signaling. Unconventional myosins modulate the spatial organization of endocytic trafficking and tether membranes or transport them along the actin cytoskeletons. Current models do not fully explain how membrane traffic forms a bridge between TGF-β and the downstream effectors that produce its functional responsiveness, such as cell migration. In this review, we present a brief overview of the current knowledge of the TGF-β signaling pathway and the molecular components that comprise the core pathway as follows: ligands, receptors, and Smad mediators. Second, we highlight key role(s) of myosin motor-mediated protein trafficking and membrane domain segregation in the modulation of the TGF-β signaling pathway. Finally, we review future challenges and provide future prospects in this field.
Collapse
|
54
|
A vivid cytokines interaction model on psoriasis with the effect of impulse biologic (TNF−αinhibitor) therapy. J Theor Biol 2019; 474:63-77. [DOI: 10.1016/j.jtbi.2019.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 11/18/2022]
|
55
|
Polanczyk MJ, Walker E, Haley D, Guerrouahen BS, Akporiaye ET. Blockade of TGF-β signaling to enhance the antitumor response is accompanied by dysregulation of the functional activity of CD4 +CD25 +Foxp3 + and CD4 +CD25 -Foxp3 + T cells. J Transl Med 2019; 17:219. [PMID: 31288845 PMCID: PMC6617864 DOI: 10.1186/s12967-019-1967-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/04/2019] [Indexed: 12/26/2022] Open
Abstract
Background The pleiotropic cytokine, transforming growth factor (TGF)-β, and CD4+CD25+Foxp3+ regulatory T cells (Tregs) play a critical role in actively suppressing antitumor immune responses. Evidence shows that TGF-β produced by tumor cells promotes tolerance via expansion of Tregs. Our group previously demonstrated that blockade of TGF-β signaling with a small molecule TGF-β receptor I antagonist (SM16) inhibited primary and metastatic tumor growth in a T cell dependent fashion. In the current study, we evaluated the effect of SM16 on Treg generation and function. Methods Using BALB/c, FoxP3eGFP and Rag−/− mice, we performed FACS analysis to determine if SM16 blocked de novo TGF-β-induced Treg generation in vitro and in vivo. CD4+ T cells from lymph node and spleen were isolated from control mice or mice maintained on SM16 diet, and flow cytometry analysis was used to detect the frequency of CD4+CD25−FoxP3+ and CD4+CD25+FoxP3+ T cells. In vitro suppression assays were used to determine the ability to suppress naive T cell proliferation in vitro of both CD4+CD25+FoxP3+ and CD4+CD25−FoxP3+ T cell sub-populations. We then examined whether SM16 diet exerted an inhibitory effect on primary tumor growth and correlated with changes in FoxP3+expression. ELISA analysis was used to measure IFN-γ levels after 72 h co-culture of CD4+CD25+ T cells from tumor-bearing mice on control or SM16 diet with CD4+CD25− T cells from naive donors. Results SM16 abrogates TGF-β-induced Treg generation in vitro but does not prevent global homeostatic expansion of CD4+ T cell sub-populations in vivo. Instead, SM16 treatment causes expansion of a population of CD4+CD25−Foxp3+ Treg-like cells without significantly altering the overall frequency of Treg in lymphoreplete naive and tumor-bearing mice. Importantly, both the CD4+CD25−Foxp3+ T cells and the CD4+CD25+Foxp3+ Tregs in mice receiving SM16 diet exhibited diminished ability to suppress naive T cell proliferation in vitro compared to Treg from mice on control diet. Conclusions These findings suggest that blockade of TGF-β signaling is a potentially useful strategy for blunting Treg function to enhance the anti-tumor response. Our data further suggest that the overall dampening of Treg function may involve the expansion of a quiescent Treg precursor population, which is CD4+CD25−Foxp3+.
Collapse
Affiliation(s)
| | - Edwin Walker
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA.,Veana Therapeutics, Inc., Portland, OR, USA
| | - Daniel Haley
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | | | - Emmanuel T Akporiaye
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA. .,Veana Therapeutics, Inc., Portland, OR, USA.
| |
Collapse
|
56
|
Silva DTD, Alves ML, Spada JCP, Silveira RDCVD, Oliveira TMFDS, Starke-Buzetti WA. Neutrophils, eosinophils, and mast cells in the intestinal wall of dogs naturally infected with Leishmania infantum. ACTA ACUST UNITED AC 2019; 27:430-438. [PMID: 30517421 DOI: 10.1590/s1984-296120180085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 10/17/2018] [Indexed: 11/22/2022]
Abstract
Visceral leishmaniasis (VL) is a disease caused by the protozoa Leishmania infantum and can cause an inflammatory reaction in the gastrointestinal tract, however the role of granulocytic cells (neutrophils, eosinophils, and mast cells) in the intestine of dogs infected is not fully understood. We performed a quantitative analysis these cells in the intestinal wall of dogs with canine visceral leishmaniasis (CVL). Twenty dogs were assigned to one of three groups: group 1 (G1, n=8), dogs with CVL and L. infantum amastigotes in the intestine; group 2 (G2, n=9), dogs with CVL but without intestinal amastigotes; and group 3 (G3, n=3), uninfected dogs (control group). Granulocytic cells were counted in the crypt-villus unit (mucosa), submucosa, and muscle layer of the intestinal mucosa. Cell counts were higher in the intestinal wall of dogs from G2 followed by G1 and G3 (p≤0.05). In G1, there was a low inverse correlation between parasite burden of the small intestine and granulocyte counts (r= -0.1, p≤0.01). However, in G2 dogs, mast cell and eosinophil numbers showed positive correlation (r=0.85, p≤0.01). The granulocytic cell hyperplasia observed in the intestine of L. infantum-infected dogs suggests that these cells may be involved in the cell-mediated immune response for parasite elimination.
Collapse
Affiliation(s)
- Diogo Tiago da Silva
- Departamento de Biologia e Ciência Animal, Faculdade de Engenharia de Ilha Solteira - FEIS, Universidade Estadual Paulista - UNESP, Ilha Solteira, SP, Brasil.,Programa de Graduação em Epidemiologia Experimental Aplicada às Zoonoses, Universidade de São Paulo - USP, São Paulo, SP, Brasil
| | - Maria Luana Alves
- Departamento de Biologia e Ciência Animal, Faculdade de Engenharia de Ilha Solteira - FEIS, Universidade Estadual Paulista - UNESP, Ilha Solteira, SP, Brasil.,Programa de Graduação em Epidemiologia Experimental Aplicada às Zoonoses, Universidade de São Paulo - USP, São Paulo, SP, Brasil
| | - Júlio Cesar Pereira Spada
- Departamento de Biologia e Ciência Animal, Faculdade de Engenharia de Ilha Solteira - FEIS, Universidade Estadual Paulista - UNESP, Ilha Solteira, SP, Brasil.,Programa de Graduação em Epidemiologia Experimental Aplicada às Zoonoses, Universidade de São Paulo - USP, São Paulo, SP, Brasil
| | | | - Trícia Maria Ferreira de Sousa Oliveira
- Programa de Graduação em Epidemiologia Experimental Aplicada às Zoonoses, Universidade de São Paulo - USP, São Paulo, SP, Brasil.,Departmento de Medicina Veterinária, Universidade de São Paulo - USP, Faculdade de Engenharia de Alimentos e Ciência Animal - FZEA, Pirassununga, SP, Brasil
| | - Wilma Aparecida Starke-Buzetti
- Departamento de Biologia e Ciência Animal, Faculdade de Engenharia de Ilha Solteira - FEIS, Universidade Estadual Paulista - UNESP, Ilha Solteira, SP, Brasil
| |
Collapse
|
57
|
Ungefroren H. Blockade of TGF-β signaling: a potential target for cancer immunotherapy? Expert Opin Ther Targets 2019; 23:679-693. [PMID: 31232607 DOI: 10.1080/14728222.2019.1636034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Malignant tumors often escape surveillance and eventual destruction by the host immune system through a variety of strategies including production of transforming growth factor (TGF)-β. Because of its generally immunosuppressive role, TGF-β has emerged as a promising therapeutic target in cancer immunotherapy. Areas covered: This article looks at specific mechanisms of how TGF-β controls the function of various immune cell subsets in the tumor microenvironment and focusses on T-cells. Various inhibition tools of TGF-β signaling and potential targets of therapeutic intervention are assessed along with the recent progress in combining TGF-β blockade and immune-mediated therapies. To round off the article, a summary of results from clinical trials is provided in which TGF-β blockade has shown therapeutic benefit for patients. Expert opinion: Data from preclinical models have shown that blocking TGF-β signaling can overcome resistance mechanisms and in combination with immune-checkpoint therapies, can yield additive or synergistic anti-tumor responses. The future of immunooncology will therefore be based on combination trials. Since response rates may critically depend on both cancer type and stage, selection of only those patients who can benefit from combinatorial immunotherapy regimens is of utmost importance.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- a First Department of Medicine , University Hospital Schleswig-Holstein, Campus Lübeck, and University of Lübeck , Lübeck , Germany.,b Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery , University Hospital Schleswig-Holstein , Campus Kiel, Kiel , Germany
| |
Collapse
|
58
|
Mendes V, Galvão I, Vieira AT. Mechanisms by Which the Gut Microbiota Influences Cytokine Production and Modulates Host Inflammatory Responses. J Interferon Cytokine Res 2019; 39:393-409. [PMID: 31013453 DOI: 10.1089/jir.2019.0011] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract encounters a wide variety of microorganisms, including beneficial symbionts, pathobionts, and pathogens. Recent evidence has shown that the gut microbiota, directly or indirectly through its components, such as metabolites, actively participates in the host inflammatory response by cytokine-microbiota or microbiota-cytokine modulation interactions, both in the gut and systemically. Therefore, further elucidation of host cytokine molecular pathways and microbiota components will provide a novel and promising therapeutic approach to control or prevent inflammatory disease and to maintain host homeostasis. The purpose of this review is to summarize well-established scientific findings and provide an updated overview regarding the direct and indirect mechanisms by which the gut microbiota can influence the inflammatory response by modulating the host's cytokine pathways that are mostly involved, but not exclusively so, with gut homeostasis. In addition, we will highlight recent results from our group, which suggest that the microbiota promotes cytokine release from inflammatory cells though activation of microbial metabolite sensor receptors that are more highly expressed on inflammatory and intestinal epithelial cells.
Collapse
Affiliation(s)
- Viviani Mendes
- 1 Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,2 Department of General Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Izabela Galvão
- 3 Department of Cellular Biology ICB, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angelica Thomaz Vieira
- 1 Laboratory of Microbiota and Immunomodulation, Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,2 Department of General Biology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
59
|
|
60
|
Buchheim JI, Matzel S, Rykova M, Vassilieva G, Ponomarev S, Nichiporuk I, Hörl M, Moser D, Biere K, Feuerecker M, Schelling G, Thieme D, Kaufmann I, Thiel M, Choukèr A. Stress Related Shift Toward Inflammaging in Cosmonauts After Long-Duration Space Flight. Front Physiol 2019; 10:85. [PMID: 30873038 PMCID: PMC6401618 DOI: 10.3389/fphys.2019.00085] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 01/24/2019] [Indexed: 01/12/2023] Open
Abstract
Space flight exerts a specific conglomerate of stressors on humans that can modulate the immune system. The mechanism remains to be elucidated and the consequences for cosmonauts in the long term are unclear. Most of the current research stems from short-term spaceflights as well as pre- and post-flight analyses due to operational limitations. Immune function of 12 cosmonauts participating in a long-duration (>140 days) spaceflight mission was monitored pre-, post-, and on two time-points in-flight. While the classical markers for stress such as cortisol in saliva where not significantly altered, blood concentrations of the endocannabinoid system (ECS) were found to be highly increased in-flight indicating a biological stress response. Moreover, subjects showed a significant rise in white blood cell counts. Neutrophils, monocytes and B cells increased by 50% whereas NK cells dropped by nearly 60% shortly after landing. Analysis of blood smears showed that lymphocyte percentages, though unchanged pre- and post-flight were elevated in-flight. Functional tests on the ground revealed stable cellular glutathione levels, unaltered baseline and stimulated ROS release in neutrophils but an increased shedding of L-selectin post-flight. In vitro stimulation of whole blood samples with fungal antigen showed a highly amplified TNF and IL-1β response. Furthermore, a significant reduction in CD4+CD25+CD27low regulatory T cells was observed post-flight but returned to normal levels after one month. Concomitantly, high in-flight levels of regulatory cytokines TGF-β, IL-10 and IL-1ra dropped rapidly after return to Earth. Finally, we observed a shift in the CD8+ T cell repertoire toward CD8+ memory cells that lasted even one month after return to Earth. Conclusion: Long-duration spaceflight triggered a sustained stress dependent release of endocannabinoids combined with an aberrant immune activation mimicking features of people at risk for inflammation related diseases. These effects persisted in part 30 days after return to Earth. The currently available repertoire of in-flight testing as well as the post-flight observation periods need to be expanded to tackle the underlying mechanism for and consequences of these immune changes in order to develop corresponding mitigation strategies based on a personalized approach for future interplanetary space explorations.
Collapse
Affiliation(s)
- Judith-Irina Buchheim
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Sandra Matzel
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Marina Rykova
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Galina Vassilieva
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Sergey Ponomarev
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Igor Nichiporuk
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Marion Hörl
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Dominique Moser
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Katharina Biere
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Matthias Feuerecker
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Gustav Schelling
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| | - Detlef Thieme
- Institute of Doping Analysis and Sports Biochemistry, Dresden, Germany
| | - Ines Kaufmann
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
- Department of Anesthesiology, Hospital Munich-Neuperlach, Munich, Germany
| | - Manfred Thiel
- Department of Anesthesiology and Surgical Intensive Care Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Choukèr
- Laboratory of Translational Research “Stress and Immunity”, Department of Anesthesiology, Hospital of the University of Munich, LMU, Munich, Germany
| |
Collapse
|
61
|
Motedayyen H, Fathi F, Fasihi-Ramandi M, Sabzghabaee AM, Taheri RA. Toll-like receptor 4 activation on human amniotic epithelial cells is a risk factor for pregnancy loss. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2019; 24:1. [PMID: 30815014 PMCID: PMC6383334 DOI: 10.4103/jrms.jrms_463_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/25/2018] [Accepted: 09/26/2018] [Indexed: 12/24/2022]
Abstract
Background: Maternal–fetal tolerance plays a fundamental role in the maintenance of pregnancy. However, this immunological tolerance can be influenced by intrauterine infections. Human amniotic epithelial cells (hAECs) have immunomodulatory effects and respond to invading pathogens through expressing various toll-like receptors (TLRs). We hypothesize that bacteria or bacterial products affect the immunosuppressive effects of hAECs through TLR stimulation. Here, we investigated how a successful pregnancy can be threatened by TLR4 activation on hAECs on lipopolysaccharide (LPS) engagement. Materials and Methods: hAECs were isolated from the amniotic membrane received from six healthy pregnant women. The immunophenotyping of hAECs was studied by flow cytometry. The isolated hAECs (4 × 105 cells/ml) were cultured in 24-well plates in the presence or absence of LPS (5 μg/ml). After 24, 48, and 72 h of incubation, the culture supernatants of hAECs were collected, and the levels of interleukin-5 (IL-5), IL-6, IL-1β, tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta 1 (TGF-β1), and prostaglandin E2 (PGE2) were measured by enzyme-linked immunosorbent assay. Results: TLR4 activation showed a stimulatory effect on TGF-β1 production of hAECs (P < 0.001–0.05). PGE2 production of LPS-stimulated hAECs was significantly increased (P < 0.01–0.05). Moreover, TLR4 could induce TNF-α and IL-1β production of hAECs (P < 0.0001–0.01), while this effect was not observed on IL-6 production of hAECs. The IL-5 was produced at a very low level in two culture supernatants of hAECs, in which its production was independent of LPS effect. Conclusion: TLR4 activation by bacterial components on hAECs may be a potential risk factor for pregnancy complications.
Collapse
Affiliation(s)
- Hossein Motedayyen
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Farshid Fathi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, System Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Mohammad Sabzghabaee
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
62
|
|
63
|
Zhao J, Qi YJ, Wang X, Jiao Y, Gong HM, Zhang JX, Jiang DY. Transforming Growth Factor-β Partially Reversed the Immunosuppressive Effect of Mesenchymal Stem Cells in Mice. Transplant Proc 2018; 50:3851-3857. [PMID: 30577277 DOI: 10.1016/j.transproceed.2018.08.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/26/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) possess powerful immunosuppression capacity. Transforming growth factor-β (TGF-β) is a well-known anti-inflammatory cytokine and plays an important role in various inflammatory processes. We hypothesized that TGF-β could synergize with MSCs in suppressing immune responses, and therefore established a mouse skin graft model to evaluate the effect of MSCs and MSCs combined with TGF-β on transplantation immunity in vivo. METHODS Balb/c and C57BL/6 mice were used to establish the skin graft model. The recipients were divided into 3 groups and received intravenous bone marrow mesenchymal stem cells (BMSCs), BMSCs pretreated with TGF-β, and 0.9% saline solution, respectively. Skin graft survival time, pathological detection, the ratio of CD4+CD25+Foxp3+Treg cell of spleens, and the level of IFN-γ, IL-2, IL-10, and TGF-β expression were tested. RESULTS The survival time of skin grafts were prolonged in both BMSC (12.5 ± 1.35 days) and BMSC-TGF-β (10.6 ± 1.90 days) recipients compared to the blank control recipients (8.0 ± 1.05 days). The ratio of CD4+CD25+Foxp3+Treg cell of spleens from BMSC and BMSC-TGF-β recipients was higher than that of the blank control, and the upregulated proliferation in the BMSC group occurred earlier and was prolonged compared to the BMSC-TGF-β group. The expression of IFN-γ and IL-2 was inhibited in both the BMSC and BMSC-TGF-β groups compared to the blank, while the expression of IL-10 and TGF-β was boosted. In contrast to the BMSC group, the BMSC-TGF-β group exhibited a weaker effect on the expression of cytokines. CONCLUSION TGF-β partially reversed the immunosuppressive effect of MSCs in vivo. This immunoregulatory feature may have potential applications for treating transplant rejection.
Collapse
Affiliation(s)
- J Zhao
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Y-J Qi
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - X Wang
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Y Jiao
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - H-M Gong
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - J-X Zhang
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China
| | - D-Y Jiang
- Department of Emergency and Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Jinan, China.
| |
Collapse
|
64
|
Yang YSS, Moynihan KD, Bekdemir A, Dichwalkar TM, Noh MM, Watson N, Melo M, Ingram J, Suh H, Ploegh H, Stellacci FR, Irvine DJ. Targeting small molecule drugs to T cells with antibody-directed cell-penetrating gold nanoparticles. Biomater Sci 2018; 7:113-124. [PMID: 30444251 PMCID: PMC6310171 DOI: 10.1039/c8bm01208c] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We sought to develop a nanoparticle vehicle that could efficiently deliver small molecule drugs to target lymphocyte populations. The synthesized amphiphilic organic ligand-protected gold nanoparticles (amph-NPs) were capable of sequestering large payloads of small molecule drugs within hydrophobic pockets of their ligand shells. These particles exhibit membrane-penetrating activity in mammalian cells, and thus enhanced uptake of a small molecule TGF-β inhibitor in T cells in cell culture. By conjugating amph-NPs with targeting antibodies or camelid-derived nanobodies, the particles' cell-penetrating properties could be temporarily suppressed, allowing targeted uptake in specific lymphocyte subpopulations. Degradation of the protein targeting moieties following particle endocytosis allowed the NPs to recover their cell-penetrating activity in situ to enter the cytoplasm of T cells. In vivo, targeted amph-NPs showed 40-fold enhanced uptake in CD8+ T cells relative to untargeted particles, and delivery of TGF-β inhibitor-loaded particles to T cells enhanced their cytokine polyfunctionality in a cancer vaccine model. Thus, this system provides a facile approach to concentrate small molecule compounds in target lymphocyte populations of interest for immunotherapy in cancer and other diseases.
Collapse
Affiliation(s)
- Yu-Sang Sabrina Yang
- Massachusetts Institute of Technology, Department of Materials Science and Engineering, Cambridge, 02139, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
TGFβ Superfamily Members as Regulators of B Cell Development and Function-Implications for Autoimmunity. Int J Mol Sci 2018; 19:ijms19123928. [PMID: 30544541 PMCID: PMC6321615 DOI: 10.3390/ijms19123928] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022] Open
Abstract
The TGFβ superfamily is composed of more than 33 growth and differentiation factors, including TGFβ1, β2, β3, BMPs, GDFs, nodal-related proteins, and activins. These members usually exert pleiotropic actions on several tissues and control multiple cellular processes, such as cell growth, cell survival, cell migration, cell fate specification, and differentiation, both during embryonic development and postnatal life. Although the effects of these factors on immune responses were elucidated long ago, most studies have been focused on the actions of TGFβs on T cells, as major regulators of adaptive immunity. In this review, we discuss new findings about the involvement of TGFβ superfamily members in the control of B cell development and function. Moreover, the potential contribution of TGFβ signaling to control B cell-mediated autoimmune diseases and its utility in the design of new therapies are also discussed.
Collapse
|
66
|
Gu S, Feng XH. TGF-β signaling in cancer. Acta Biochim Biophys Sin (Shanghai) 2018; 50:941-949. [PMID: 30165534 DOI: 10.1093/abbs/gmy092] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 07/16/2018] [Indexed: 12/19/2022] Open
Abstract
Signals from the transforming growth factor-β (TGF-β) superfamily mediate a broad spectrum of cellular processes and are deregulated in many diseases, including cancer. TGF-β signaling has dual roles in tumorigenesis. In the early phase of tumorigenesis, TGF-β has tumor suppressive functions, primarily through cell cycle arrest and apoptosis. However, in the late stage of cancer, TGF-β acts as a driver of tumor progression and metastasis by increasing tumor cell invasiveness and migration and promoting chemo-resistance. Here, we briefly review the mechanisms and functions of TGF-β signaling during tumor progression and discuss the therapeutic potentials of targeting the TGF-β pathway in cancer.
Collapse
Affiliation(s)
- Shuchen Gu
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
67
|
Choi B, Kim SH. Regulatory T Cells Promote Pancreatic Islet Function and Viability via TGF-β1 in vitro and in vivo. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2018. [DOI: 10.15324/kjcls.2018.50.3.304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Bongkum Choi
- Transplantation Research Center, Clinical Research Institute, Samsung Biomedical Research Institute, Seoul, Korea
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sa-Hyun Kim
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| |
Collapse
|
68
|
The effect of lipopolysaccharide on anti-inflammatory and pro-inflammatory cytokines production of human amniotic epithelial cells. Reprod Biol 2018; 18:404-409. [PMID: 30220549 DOI: 10.1016/j.repbio.2018.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/17/2018] [Accepted: 09/08/2018] [Indexed: 12/18/2022]
Abstract
Intrauterine infection is a major cause of immune imbalance at the maternal-fetal interface, which leads to spontaneous abortion, premature rupture of the fetal membranes, and preterm birth. Human amniotic epithelial cells (hAECs) play a fundamental role in the maintenance of pregnancy. We hypothesize that bacteria influence the immunomodulatory effects of hAECs through stimulation of Toll-like receptors (TLRs). Here, we investigated how lipopolysaccharide (LPS) as a bacterial component affects anti-inflammatory and pro-inflammatory cytokines production of hAECs. Human placentas were obtained from six healthy pregnant women and hAECs were isolated. The phenotypic characteristics of hAECs were determined by flow cytometry. The hAECs (4 × 105 cells/ml) were cultured in the presence or absence of LPS (5 μg/ml). The viability of the cells was assessed and culture supernatants of hAECs were collected after 24, 48 and 72 h of incubation. The levels of transforming growth factor-beta1 (TGF-β1), interleukin-4 (IL-4), tumor necrosis factor-alpha (TNF-α), interleukin-17 A (IL-17A), and interferon-gamma (IFN-γ) were measured by ELISA. Our data showed that LPS treatment did not affect the viability of hAECs, while had a stimulatory effect on TGF-β1 production of hAECs (p < 0.001). A significant reduction in IL-4 production of LPS-stimulated hAECs was observed (p < 0.05). LPS enhanced the production of TNF-α and IL-17 A of hAECs (p < 0.05-0.0001). The IFN-γ level was only detectable in two culture supernatants of hAECs, and the level was unchanged after stimulation with LPS. Based on these findings, LPS may play a pivotal role in immune imbalance at the feto-maternal interface through affecting anti-inflammatory and pro-inflammatory cytokines production of hAECs.
Collapse
|
69
|
Marino M, Pontrelli G, Vairo G, Wriggers P. A chemo-mechano-biological formulation for the effects of biochemical alterations on arterial mechanics: the role of molecular transport and multiscale tissue remodelling. J R Soc Interface 2018; 14:rsif.2017.0615. [PMID: 29118114 DOI: 10.1098/rsif.2017.0615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
This paper presents a chemo-mechano-biological framework for arterial physiopathology. The model accounts for the fine remodelling in the multiscale hierarchical arrangement of tissue constituents and for the diffusion of molecular species involved in cell-cell signalling pathways. Effects in terms of alterations in arterial compliance are obtained. A simple instructive example is introduced. Although oversimplified with respect to realistic case studies, the proposed application mimics the biochemical activity of matrix metalloproteinases, transforming growth factors beta and interleukins on tissue remodelling. Effects of macrophage infiltration, of intimal thickening and of a healing phase are investigated, highlighting the corresponding influence on arterial compliance. The obtained results show that the present approach is able to capture changes in arterial mechanics as a consequence of the alterations in tissue biochemical environment and cellular activity, as well as to incorporate the protective role of both autoimmune responses and pharmacological treatments.
Collapse
Affiliation(s)
- Michele Marino
- Institut für Kontinuumsmechanik, Leibniz Universität Hannover, Hannover, Germany
| | - Giuseppe Pontrelli
- Istituto per le Applicazioni del Calcolo, National Research Council (CNR), Rome, Italy
| | - Giuseppe Vairo
- Dipartimento di Ingegneria Civile e Ingegneria Informatica, Università degli Studi di Roma 'Tor Vergata', Rome, Italy
| | - Peter Wriggers
- Institut für Kontinuumsmechanik, Leibniz Universität Hannover, Hannover, Germany
| |
Collapse
|
70
|
Abstract
Transforming Growth Factor beta (TGF-β) is a pleiotropic cytokine produced in large amounts within cancer microenvironments that will ultimately promote neoplastic progression, notably by suppressing the host’s T-cell immunosurveillance. This effect is mostly due to the well-known inhibitory effect of TGF-β on T cell proliferation, activation, and effector functions. Moreover, TGF-β subverts T cell immunity by favoring regulatory T-cell differentiation, further reinforcing immunosuppression within tumor microenvironments. These findings stimulated the development of many strategies to block TGF-β or its signaling pathways, either as monotherapy or in combination with other therapies, to restore anti-cancer immunity. Paradoxically, recent studies provided evidence that TGF-β can also promote differentiation of certain inflammatory populations of T cells, such as Th17, Th9, and resident-memory T cells (Trm), which have been associated with improved tumor control in several models. Here, we review current advances in our understanding of the many roles of TGF-β in T cell biology in the context of tumor immunity and discuss the possibility to manipulate TGF-β signaling to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Amina Dahmani
- Centre de Recherche de L'hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, QC H1T 2M4, Canada.
| | - Jean-Sébastien Delisle
- Centre de Recherche de L'hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, QC H1T 2M4, Canada.
- Hematology-Oncology service, Hôpital Maisonneuve-Rosemont, Department of Medicine, Université de Montréal, Montréal, QC H1T 2M4, Canada.
| |
Collapse
|
71
|
TGF-β in T Cell Biology: Implications for Cancer Immunotherapy. Cancers (Basel) 2018; 10:cancers10060194. [PMID: 29891791 PMCID: PMC6025055 DOI: 10.3390/cancers10060194] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 12/25/2022] Open
Abstract
Transforming Growth Factor beta (TGF-β) is a pleiotropic cytokine produced in large amounts within cancer microenvironments that will ultimately promote neoplastic progression, notably by suppressing the host’s T-cell immunosurveillance. This effect is mostly due to the well-known inhibitory effect of TGF-β on T cell proliferation, activation, and effector functions. Moreover, TGF-β subverts T cell immunity by favoring regulatory T-cell differentiation, further reinforcing immunosuppression within tumor microenvironments. These findings stimulated the development of many strategies to block TGF-β or its signaling pathways, either as monotherapy or in combination with other therapies, to restore anti-cancer immunity. Paradoxically, recent studies provided evidence that TGF-β can also promote differentiation of certain inflammatory populations of T cells, such as Th17, Th9, and resident-memory T cells (Trm), which have been associated with improved tumor control in several models. Here, we review current advances in our understanding of the many roles of TGF-β in T cell biology in the context of tumor immunity and discuss the possibility to manipulate TGF-β signaling to improve cancer immunotherapy.
Collapse
|
72
|
Holmgaard RB, Schaer DA, Li Y, Castaneda SP, Murphy MY, Xu X, Inigo I, Dobkin J, Manro JR, Iversen PW, Surguladze D, Hall GE, Novosiadly RD, Benhadji KA, Plowman GD, Kalos M, Driscoll KE. Targeting the TGFβ pathway with galunisertib, a TGFβRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J Immunother Cancer 2018; 6:47. [PMID: 29866156 PMCID: PMC5987416 DOI: 10.1186/s40425-018-0356-4] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 05/11/2018] [Indexed: 12/12/2022] Open
Abstract
Background TGFβ signaling plays a pleotropic role in tumor biology, promoting tumor proliferation, invasion and metastasis, and escape from immune surveillance. Inhibiting TGFβ’s immune suppressive effects has become of particular interest as a way to increase the benefit of cancer immunotherapy. Here we utilized preclinical models to explore the impact of the clinical stage TGFβ pathway inhibitor, galunisertib, on anti-tumor immunity at clinically relevant doses. Results In vitro treatment with galunisertib reversed TGFβ and regulatory T cell mediated suppression of human T cell proliferation. In vivo treatment of mice with established 4T1-LP tumors resulted in strong dose-dependent anti-tumor activity with close to 100% inhibition of tumor growth and complete regressions upon cessation of treatment in 50% of animals. This effect was CD8+ T cell dependent, and led to increased T cell numbers in treated tumors. Mice with durable regressions rejected tumor rechallenge, demonstrating the establishment of immunological memory. Consequently, mice that rejected immunogenic 4T1-LP tumors were able to resist rechallenge with poorly immunogenic 4 T1 parental cells, suggesting the development of a secondary immune response via antigen spreading as a consequence of effective tumor targeting. Combination of galunisertib with PD-L1 blockade resulted in improved tumor growth inhibition and complete regressions in colon carcinoma models, demonstrating the potential synergy when cotargeting TGFβ and PD-1/PD-L1 pathways. Combination therapy was associated with enhanced anti-tumor immune related gene expression profile that was accelerated compared to anti-PD-L1 monotherapy. Conclusions Together these data highlight the ability of galunisertib to modulate T cell immunity and the therapeutic potential of combining galunisertib with current PD-1/L1 immunotherapy.
Collapse
Affiliation(s)
- Rikke B Holmgaard
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - David A Schaer
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Yanxia Li
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Stephen P Castaneda
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Mary Y Murphy
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Xiaohong Xu
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Ivan Inigo
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Julie Dobkin
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Jason R Manro
- 0000 0000 2220 2544grid.417540.3Lilly Research Laboratories, Eli Lilly and Company 46285 Indianapolis IN USA
| | - Philip W Iversen
- 0000 0000 2220 2544grid.417540.3Lilly Research Laboratories, Eli Lilly and Company 46285 Indianapolis IN USA
| | - David Surguladze
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Gerald E Hall
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Ruslan D Novosiadly
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Karim A Benhadji
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Gregory D Plowman
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| | - Michael Kalos
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA.,Janssen Pharmaceutical Companies of Johnson and Johnson Spring House PA USA
| | - Kyla E Driscoll
- 0000 0000 2220 2544grid.417540.3Eli Lilly and Company 450 East 29th Street New York USA
| |
Collapse
|
73
|
Liu T, Huang J, Li Y, Ehsan M, Wang S, Zhou Z, Song X, Yan R, Xu L, Li X. Molecular characterisation and the protective immunity evaluation of Eimeria maxima surface antigen gene. Parasit Vectors 2018; 11:325. [PMID: 29848353 PMCID: PMC5977735 DOI: 10.1186/s13071-018-2906-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/20/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Coccidiosis is recognised as a major parasitic disease in chickens. Eimeria maxima is considered as a highly immunoprotective species within the Eimeria spp. family that infects chickens. In the present research, the surface antigen gene of E. maxima (EmSAG) was cloned, and the ability of EmSAG to stimulate protection against E. maxima was evaluated. METHODS Prokaryotic and eukaryotic plasmids expressing EmSAG were constructed. The EmSAG transcription and expression in vivo was performed based on the RT-PCR and immunoblot analysis. The expression of EmSAG in sporozoites and merozoites was detected through immunofluorescence analyses. The immune protection was assessed based on challenge experiments. Flow cytometry assays were used to determine the T cell subpopulations. The serum antibody and cytokine levels were evaluated by ELISA. RESULTS The open reading frame (ORF) of EmSAG gene contained 645 bp encoding 214 amino acid residues. The immunoblot and RT-PCR analyses indicated that the EmSAG gene were transcribed and expressed in vivo. The EmSAG proteins were expressed in sporozoite and merozoite stages of E. maxima by the immunofluorescence assay. Challenge experiments showed that both pVAX1-SAG and the recombinant EmSAG (rEmSAG) proteins were successful in alleviating jejunal lesions, decreasing loss of body weight and the oocyst ratio. Additionally, these experiments possessed anticoccidial indices (ACI) of more than 170. Higher percentages of CD4+ and CD8+ T cells were detected in both EmSAG-inoculated birds than those of the negative control groups (P < 0.05). The EmSAG-specific antibody concentrations of both the rEmSAG and pVAX1-EmSAG groups were much higher than those of the negative controls (P < 0.05). Higher concentrations of IL-4, IFN-γ, TGF-β1 and IL-17 were observed more in both the rEmSAG protein and pVAX1-SAG inoculated groups than those of negative controls (P < 0.05). CONCLUSIONS Our findings suggest that EmSAG is capable of eliciting a moderate immune protection and could be used as an effective vaccine candidate against E. maxima.
Collapse
Affiliation(s)
- Tingqi Liu
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Jingwei Huang
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Yanlin Li
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Muhammad Ehsan
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Shuai Wang
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Zhouyang Zhou
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Xiaokai Song
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Ruofeng Yan
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Lixin Xu
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China
| | - Xiangrui Li
- College of Veterinary Medicine, Nanjing Agriculture University, 1 Weigang, Nanjing, Jiangsu, 210095, People's Republic of China.
| |
Collapse
|
74
|
Human amniotic epithelial cells inhibit activation and pro-inflammatory cytokines production of naive CD4+ T cells from women with unexplained recurrent spontaneous abortion. Reprod Biol 2018; 18:182-188. [PMID: 29729842 DOI: 10.1016/j.repbio.2018.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 04/26/2018] [Indexed: 01/27/2023]
Abstract
Unexplained recurrent spontaneous abortion (URSA) has been assumed to be caused by a defect in maternal immunological tolerance to the fetus. Human amniotic epithelial cells (hAECs) have stem cell-like features and the ability to modulate the innate and adoptive immune responses. This study aimed to investigate whether hAECs have immunomodulatory effects on naive CD4+ T cells from URSA patients. hAECs were obtained from 15 healthy pregnant women and phenotypic profile of hAECs was determined by flow cytometry. Naive CD4+ T cells were isolated from 25 URSA patients using an immunomagnetic separation method. Naive T cells were stimulated with anti-CD3/anti-CD28 antibodies and co-cultured with different numbers of hAECs for 3 and 6 days. Immunomodulatory effect of hAECs on activation of stimulated T cell was assessed by flow cytometry and Enzyme-linked immunoasorbent assay (ELISA). The hAECs effect on pro-inflammatory cytokines production of activated T cells was also measured by ELISA. Our results indicated that hAECs significantly inhibited the activation of naive T cells in a dose-dependent manner (p < 0.0001-0.05). They significantly reduced the production of transforming growth factor-beta1 (TGF-β1) of stimulated CD4+T cells (p < 0.0001-0.05). Moreover, hAECs had potent immunomodulatory effects on the production of interferon-gamma (IFN-γ) and interleukin-17A (IL-17A) of activated T cells (p < 0.0001-0.01). These findings suggest that hAECs may be a suitable cell source to modulate abnormal immune responses in women with URSA.
Collapse
|
75
|
Tsutsumi-Kuroda U, Inoue T, Futakuchi A, Shobayashi K, Takahashi E, Kojima S, Inoue-Mochita M, Fujimoto T, Tanihara H. Decreased MCP-1/CCR2 axis-mediated chemotactic effect of conjunctival fibroblasts after transdifferentiation into myofibroblasts. Exp Eye Res 2018; 170:76-80. [DOI: 10.1016/j.exer.2018.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 01/19/2018] [Accepted: 02/12/2018] [Indexed: 02/03/2023]
|
76
|
Caslin HL, Kiwanuka KN, Haque TT, Taruselli MT, MacKnight HP, Paranjape A, Ryan JJ. Controlling Mast Cell Activation and Homeostasis: Work Influenced by Bill Paul That Continues Today. Front Immunol 2018; 9:868. [PMID: 29755466 PMCID: PMC5932183 DOI: 10.3389/fimmu.2018.00868] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/09/2018] [Indexed: 01/13/2023] Open
Abstract
Mast cells are tissue resident, innate immune cells with heterogenous phenotypes tuned by cytokines and other microenvironmental stimuli. Playing a protective role in parasitic, bacterial, and viral infections, mast cells are also known for their role in the pathogenesis of allergy, asthma, and autoimmune diseases. Here, we review factors controlling mast cell activation, with a focus on receptor signaling and potential therapies for allergic disease. Specifically, we will discuss our work with FcεRI and FγR signaling, IL-4, IL-10, and TGF-β1 treatment, and Stat5. We conclude with potential therapeutics for allergic disease. Much of these efforts have been influenced by the work of Bill Paul. With many mechanistic targets for mast cell activation and different classes of therapeutics being studied, there is reason to be hopeful for continued clinical progress in this area.
Collapse
Affiliation(s)
- Heather L Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Kasalina N Kiwanuka
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Tamara T Haque
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Marcela T Taruselli
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - H Patrick MacKnight
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Anuya Paranjape
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
77
|
Botti C, Seregni E, Ferrari L, Martinetti A, Bombardieri E. Immunosuppressive Factors: Role in Cancer Development and Progression. Int J Biol Markers 2018; 13:51-69. [PMID: 9803353 DOI: 10.1177/172460089801300201] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The concept of the immunological surveillance against neoplastic cells was initially proposed by Erlich in 1909 and later elaborated by Burnet. This hypothesis states that the normal function of the immune system, in particular the cell-mediated immunity, is to recognize and destroy the transformed and proliferating tumor cells. The role of cell-mediated immunity during the first steps of tumorigenesis remains controversial. However, there is certain evidence about its importance in the progression and dissemination of cancer. The frequent immunosuppressed condition of cancer patients at tumor relapse or recurrence of secondary tumors is a clinical sign supporting this hypothesis, and many studies have demonstrated a defective immune response in patients diagnosed with advanced cancer. Several mechanisms of escape from the immune surveillance have been described, including the immunoselection of tumor antigen-negative variants, the downregulation of MHC class I expression, suppressive T cells, and the elaboration of immunosuppressive cytokines and other factors. Because of the technical difficulty of isolating the very small amounts from culture supernatants or body fluids, only a few of these substances have been characterized and studied with respect to their biological activity: transforming growth factor-β (TGF-β), the protein p15E, interleukin 10 (IL-10), prostaglandin E2 (PGE2), mucins, suppressive E-receptor (SER), immunosuppressive acidic protein (IAP), and adhesion molecules. The possibility of monitoring cancer patients by testing biochemical factors related to cancer growth led to a proposal to measure a number of these factors as tumor markers. Some of them, e.g mucins, enjoy the consensus of the oncologic community, as for some indications they can help the clinician in the management of cancer patients. Except for the class of mucins, the other above-mentioned immunosuppressive factors have not found any clinical application in the laboratory routine because the information deriving from their measurement, although of speculative and scientific interest, has limited clinical value at present. Nevertheless, even if they have no impact on patient management, these substances do have a potential role to play in the study of cancer patients, and should be taken into account when developing new therapeutic strategies.
Collapse
Affiliation(s)
- C Botti
- Nuclear Medicine Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milano, Italy
| | | | | | | | | |
Collapse
|
78
|
Reyes-Cerpa S, Vallejos-Vidal E, Gonzalez-Bown MJ, Morales-Reyes J, Pérez-Stuardo D, Vargas D, Imarai M, Cifuentes V, Spencer E, Sandino AM, Reyes-López FE. Effect of yeast (Xanthophyllomyces dendrorhous) and plant (Saint John's wort, lemon balm, and rosemary) extract based functional diets on antioxidant and immune status of Atlantic salmon (Salmo salar) subjected to crowding stress. FISH & SHELLFISH IMMUNOLOGY 2018; 74:250-259. [PMID: 29305990 DOI: 10.1016/j.fsi.2017.12.061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 12/18/2017] [Accepted: 12/31/2017] [Indexed: 06/07/2023]
Abstract
Salmon farming may face stress due to the intensive culture conditions with negative impacts on overall performance. In this aspect, functional feed improves not only the basic nutritional requirements but also the health status and fish growth. However, to date no studies have been carried out to evaluate the effect of functional diets in salmon subjected to crowding stress. Thus, the aim of this study was to evaluate the effect of yeast extract (Xanthophyllomyces dendrorhous; diet A) and the combination of plant extracts (common Saint John's wort, lemon balm, and rosemary; diet B) on the antioxidant and immune status of Atlantic salmon grown under normal cultured conditions and then subjected to crowding stress. Fish were fed with functional diets during 30 days (12 kg/m3) and then subjected to crowding stress (20 kg/m3) for 10 days. The lipid peroxidation in gut showed that both diets induced a marked decrease on oxidative damage when fish were subjected to crowding stress. The protein carbonylation in muscle displayed at day 30 a marked decrease in both functional diets that was more marked on the stress condition. The expression of immune markers (IFNγ, CD4, IL-10, TGF-β, IgMmb, IgMsec, T-Bet, and GATA-3) indicated the upregulation of those associated to humoral-like response (CD4, IL-10, GATA-3) when fish were subjected to crowding stress. These results were confirmed with the expression of secreted IgM. Altogether, these functional diets improved the antioxidant status and increased the expression of genes related to Th2-like response suggesting a protective role on fish subjected to crowding stress.
Collapse
Affiliation(s)
| | - Eva Vallejos-Vidal
- Department of Cell Biology, Physiology and Immunology, Faculty of Biosciences (Building C), Universitat Autonoma de Barcelona, 08193, Bellaterra, Spain
| | - María José Gonzalez-Bown
- Laboratorio de Virología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Jonathan Morales-Reyes
- Laboratorio de Virología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Diego Pérez-Stuardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Chile
| | - Deborah Vargas
- Laboratorio de Virología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Mónica Imarai
- Laboratorio de Inmunología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Víctor Cifuentes
- Laboratorio de Genética, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Eugenio Spencer
- Laboratorio de Virología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Ana María Sandino
- Laboratorio de Virología, Centro de Biotecnología Acuícola, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile; ActivaQ S.A. General del Canto 460, Providencia, Santiago, Chile.
| | - Felipe E Reyes-López
- Department of Cell Biology, Physiology and Immunology, Faculty of Biosciences (Building C), Universitat Autonoma de Barcelona, 08193, Bellaterra, Spain.
| |
Collapse
|
79
|
Oncolytic adenovirus coexpressing interleukin-12 and decorin overcomes Treg-mediated immunosuppression inducing potent antitumor effects in a weakly immunogenic tumor model. Oncotarget 2018; 8:4730-4746. [PMID: 28002796 PMCID: PMC5354867 DOI: 10.18632/oncotarget.13972] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 12/01/2016] [Indexed: 01/02/2023] Open
Abstract
Interleukin (IL)-12 is a potent antitumor cytokine. However, immunosuppressive tumor microenvironments containing transforming growth factor-β (TGF-β) attenuate cytokine-mediated antitumor immune responses. To enhance the efficacy of IL-12-mediated cancer immunotherapy, decorin (DCN) was explored as an adjuvant for overcoming TGF-β-mediated immunosuppression. We designed and generated a novel oncolytic adenovirus (Ad) coexpressing IL-12 and DCN (RdB/IL12/DCN). RdB/IL12/DCN-treated tumors showed significantly greater levels of interferon (IFN)-γ, tumor necrosis factor-α, monocyte chemoattractant protein-1, and IFN-γ-secreting immune cells than tumors treated with cognate control oncolytic Ad expressing a single therapeutic gene (RdB/DCN or RdB/IL12). Moreover, RdB/IL12/DCN attenuated intratumoral TGF-β expression, which positively correlated with reduction of Treg cells in draining lymph nodes and tumor tissues. Furthermore, tumor tissue treated with RdB/IL12/DCN showed increases infiltration of CD8+ T cells and proficient viral spreading within tumor tissues. These results demonstrated that an oncolytic Ad co-expressing IL-12 and DCN induces a potent antitumor immune response via restoration of antitumor immune function in a weakly immunogenic murine 4T1 orthotopic breast cancer model. These findings provide new insights into the therapeutic mechanisms of IL-12 plus DCN, making it a promising cancer immunotherapeutic agent for overcoming tumor-induced immunosuppression.
Collapse
|
80
|
The emerging roles of exosomes in leukemogeneis. Oncotarget 2018; 7:50698-50707. [PMID: 27191983 PMCID: PMC5226614 DOI: 10.18632/oncotarget.9333] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Accepted: 05/05/2016] [Indexed: 12/21/2022] Open
Abstract
Communication between leukemia cells and their environment is essential for the development and progression of leukemia. Exosomes are microvesicles secreted by many types of cells that contain protein and RNA and mediate intercellular communication. The involvement of exosomes has been demonstrated in the crosstalk between leukemic cells, stromal cells and endothelial cells, consequently promoting the survival of leukemic cells, protection of leukemic cells from the cytotoxic effects of chemotherapeutic drugs, angiogenesis and cell migration. At the same time, exosomes can be used for the detection and monitoring of leukemia, with some advantage over current methods of detection and surveillance. As they are involved in immune response towards leukemic cells, exosomes can also potentially be exploited to augment immunotherapy in leukemia. In this review, we first describe the general characteristics of exosomes and biogenesis of exosomes. We then highlight the emerging role of exosomes in different types of leukemia. Finally, the clinical value of exosomes as biomarkers, in vivo drug carriers and novel exosome-based immunotherapy are discussed.
Collapse
|
81
|
DeLong JH, Hall AO, Konradt C, Coppock GM, Park J, Harms Pritchard G, Hunter CA. Cytokine- and TCR-Mediated Regulation of T Cell Expression of Ly6C and Sca-1. THE JOURNAL OF IMMUNOLOGY 2018; 200:1761-1770. [PMID: 29358280 DOI: 10.4049/jimmunol.1701154] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/21/2017] [Indexed: 02/05/2023]
Abstract
Ly6C and Sca-1 (Ly6A/E) are Ly6 family GPI-anchored surface molecules that are differentially expressed by multiple immune populations. Ly6C expression has been used to distinguish short-lived effector CD4+ T cells from memory precursor effector cells, whereas Sca-1 has been used in the identification of CD8+ memory stem cells. This study examines the expression patterns of these molecules and establishes that, in vitro, IL-27, type I IFN, and IFN-γ are potent inducers of Ly6C and Sca-1 in naive mouse CD4+ and CD8+ T cells, whereas TGF-β limits their expression. The induction of Ly6C and Sca-1 by IL-27 and IFN-γ is dependent on STAT1, but not STAT3 or T-bet. In mouse splenocytes, at homeostasis, Ly6C and Sca-1 expression was not restricted to effector cells, but was also found at various levels on naive and memory populations. However, in response to infection with Toxoplasma gondii, pathogen-specific T cells expressed high levels of these molecules and in this context, endogenous IL-27 and IFN-γ were required for the expression of Ly6C but not Sca-1. Together, these findings highlight the TCR-dependent and cytokine-mediated signals that modulate T cell expression of Ly6C and Sca-1 in vitro and in vivo during infection.
Collapse
Affiliation(s)
- Jonathan H DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Aisling O'Hara Hall
- Immunology Discovery Research, Janssen Research and Development, LLC, Spring House, PA 19002
| | - Christoph Konradt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Gaia M Coppock
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Department of Nephrology, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Jeongho Park
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | | | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
82
|
Warsinske HC, Pienaar E, Linderman JJ, Mattila JT, Kirschner DE. Deletion of TGF-β1 Increases Bacterial Clearance by Cytotoxic T Cells in a Tuberculosis Granuloma Model. Front Immunol 2017; 8:1843. [PMID: 29326718 PMCID: PMC5742530 DOI: 10.3389/fimmu.2017.01843] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis is the pathogenic bacterium that causes tuberculosis (TB), one of the most lethal infectious diseases in the world. The only vaccine against TB is minimally protective, and multi-drug resistant TB necessitates new therapeutics to treat infection. Developing new therapies requires a better understanding of the complex host immune response to infection, including dissecting the processes leading to formation of granulomas, the dense cellular lesions associated with TB. In this work, we pair experimental and computational modeling studies to explore cytokine regulation in the context of TB. We use our next-generation hybrid multi-scale model of granuloma formation (GranSim) to capture molecular, cellular, and tissue scale dynamics of granuloma formation. We identify TGF-β1 as a major inhibitor of cytotoxic T-cell effector function in granulomas. Deletion of TGF-β1 from the system results in improved bacterial clearance and lesion sterilization. We also identify a novel dichotomous regulation of cytotoxic T cells and macrophages by TGF-β1 and IL-10, respectively. These findings suggest that increasing cytotoxic T-cell effector functions may increase bacterial clearance in granulomas and highlight potential new therapeutic targets for treating TB.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elsje Pienaar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
83
|
Bajnok A, Berta L, Orbán C, Veres G, Zádori D, Barta H, Méder Ü, Vécsei L, Tulassay T, Szabó M, Toldi G. Distinct cytokine patterns may regulate the severity of neonatal asphyxia-an observational study. J Neuroinflammation 2017; 14:244. [PMID: 29233180 PMCID: PMC5727967 DOI: 10.1186/s12974-017-1023-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/05/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation and a systemic inflammatory reaction are important features of perinatal asphyxia. Neuroinflammation may have dual aspects being a hindrance, but also a significant help in the recovery of the CNS. We aimed to assess intracellular cytokine levels of T-lymphocytes and plasma cytokine levels in moderate and severe asphyxia in order to identify players of the inflammatory response that may influence patient outcome. METHODS We analyzed the data of 28 term neonates requiring moderate systemic hypothermia in a single-center observational study. Blood samples were collected between 3 and 6 h of life, at 24 h, 72 h, 1 week, and 1 month of life. Neonates were divided into a moderate (n = 17) and a severe (n = 11) group based on neuroradiological and amplitude-integrated EEG characteristics. Peripheral blood mononuclear cells were assessed with flow cytometry. Cytokine plasma levels were measured using Bioplex immunoassays. Components of the kynurenine pathway were assessed by high-performance liquid chromatography. RESULTS The prevalence and extravasation of IL-1b + CD4 cells were higher in severe than in moderate asphyxia at 6 h. Based on Receiver operator curve analysis, the assessment of the prevalence of CD4+ IL-1β+ and CD4+ IL-1β+ CD49d+ cells at 6 h appears to be able to predict the severity of the insult at an early stage in asphyxia. Intracellular levels of TNF-α in CD4 cells were increased at all time points compared to 6 h in both groups. At 1 month, intracellular levels of TNF-α were higher in the severe group. Plasma IL-6 levels were higher at 1 week in the severe group and decreased by 1 month in the moderate group. Intracellular levels of IL-6 peaked at 24 h in both groups. Intracellular TGF-β levels were increased from 24 h onwards in the moderate group. CONCLUSIONS IL-1β and IL-6 appear to play a key role in the early events of the inflammatory response, while TNF-α seems to be responsible for prolonged neuroinflammation, potentially contributing to a worse outcome. The assessment of the prevalence of CD4+ IL-1β+ and CD4+ IL-1β+ CD49d+ cells at 6 h appears to be able to predict the severity of the insult at an early stage in asphyxia.
Collapse
Affiliation(s)
- Anna Bajnok
- First Department of Obstetrics and Gynecology, Semmelweis University, Baross str. 27, Budapest, H-1088, Hungary.,First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - László Berta
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Csaba Orbán
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Gábor Veres
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Dénes Zádori
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary
| | - Hajnalka Barta
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - Ünőke Méder
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Clinical Centre, Faculty of Medicine, University of Szeged, Semmelweis str. 6, 5th floor, Szeged, H-6725, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| | - Tivadar Tulassay
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Miklós Szabó
- First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary.,MTA-SE Pediatrics and Nephrology Research Group, Budapest, Hungary
| | - Gergely Toldi
- First Department of Obstetrics and Gynecology, Semmelweis University, Baross str. 27, Budapest, H-1088, Hungary. .,First Department of Pediatrics, Semmelweis University, Bókay János str. 53-54, Budapest, H-1083, Hungary. .,Birmingham Women's and Children's Hospital, Neonatal Unit, Birmingham, UK.
| |
Collapse
|
84
|
Inhibition of T Cell Alloreactivity by Bronchial Epithelium Is Impaired in Lung Transplant Recipients, Through Pathways Involving TGF-β, IL-10 and HLA-G. Transplantation 2017; 101:2192-2199. [PMID: 27820781 DOI: 10.1097/tp.0000000000001553] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) after lung transplantation (LTx) results from bronchial epithelial cell (BECs) damages, thought to be orchestrated by T cells primed by antigen-presenting cell presenting alloantigens. In this cell cross-talk, BECs are also suspected to play a pivotal immunosuppressive role in T cell alloreactivity. We studied the immunomodulating role of BECs in a human ex vivo model of allogeneic T cell response, both in healthy subjects and LTx recipients. METHODS BECs from 35 LTx recipients (n = 22 stable, n = 13 BOS) and healthy controls (n = 25) were cultured as primary cell cultures. Their inhibitory capacities through the involvement of tolerogenic molecules (HLA-G, TGF-β, and IL-10) were tested on a mixed lymphocyte reaction between antigen-presenting cells and recipient T cells. RESULTS Control BECs inhibited T cell alloproliferation by a mean of 53 ± 7%. This inhibitory effect of BECs was significantly reduced in the stable LTx group (24 ± 8%, P = 0.009), but not in the BOS TxP group (53 ± 10%, P = 0.97). Neutralization of HLA-G, TGF-β, and IL-10 partially restored T cell alloproliferation, arguing for their involvement in the immunosuppressive effect of BECs. BECs culture supernatant from stable LTx patients with impaired BEC properties showed a skewed Th2-type secretion profile (high IL-4/IFN-γ ratio). CONCLUSIONS The inhibitory properties of BECs are dysregulated in stable LTx recipients, which could suggest their instrumental role in the initiation of BOS process and potential targeted therapies.
Collapse
|
85
|
Röhe I, Göbel TW, Goodarzi Boroojeni F, Zentek J. Effect of feeding soybean meal and differently processed peas on the gut mucosal immune system of broilers. Poult Sci 2017; 96:2064-2073. [PMID: 28339726 DOI: 10.3382/ps/pew491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 12/19/2016] [Indexed: 11/20/2022] Open
Abstract
Peas are traditionally used as a protein source for poultry. However, peas contain antinutritional factors (ANF), which are associated with the initiation of local and systemic immune reactions. The current study examined the effect of feeding raw or differently processed peas in comparison with feeding a soybean meal (SBM) based control diet (C) on the gut mucosal immune system of broilers in a 35 day feeding trial. In six replicates, a total of 360 one-day-old male broilers were randomly allocated to four different groups receiving C, or three treatment diets containing raw, fermented, and enzymatically pre-digested peas, each supplying 30% of required crude protein. After slaughtering, jejunal samples were taken for immunohistochemical, flow cytometric, and gene expression analyses. Investigations were focused on the topological distribution of intraepithelial leukocytes (villus tip, villus mid, and crypt region) as well as on the further characterization of the different intraepithelial lymphocytes (IEL) and concomitant pro- and anti-inflammatory cytokines. Broilers receiving the raw or processed pea diets had higher numbers of intraepithelial CD45+ leukocytes in the tip (P = 0.004) and mid region (P < 0.001) of villi than birds fed C. Higher numbers of intraepithelial CD3+ lymphocytes were found in the villus tip (P = 0.002) and mid region (P = 0.003) of birds fed raw or processed pea containing diets in comparison with those fed C. The flow cytometric phenotyping showed a similar relative distribution of IEL among the feeding groups. The expression of intestinal pro- and anti-inflammatory cytokines was affected by feeding the different diets only to a minor extent. To conclude, feeding of diets formulated with raw and processed peas in comparison with feeding a SBM control diet initiated mucosal immune responses in the jejunum of broilers indicated by a quantitative increase of intraepithelial T cells. Further research is needed in order to ascertain the specific factors which are responsible for observed local immune reactions and how these local reactions might affect the immune status and health of broilers.
Collapse
Affiliation(s)
- I Röhe
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany
| | - T W Göbel
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstr 13, 80539 Munich, Germany
| | - F Goodarzi Boroojeni
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany
| | - J Zentek
- Institute of Animal Nutrition, Department of Veterinary Medicine, Freie Universität Berlin, Königin-Luise-Str. 49, 14195 Berlin, Germany
| |
Collapse
|
86
|
Thomas BJ, Kan-O K, Loveland KL, Elias JA, Bardin PG. In the Shadow of Fibrosis: Innate Immune Suppression Mediated by Transforming Growth Factor-β. Am J Respir Cell Mol Biol 2017; 55:759-766. [PMID: 27603223 DOI: 10.1165/rcmb.2016-0248ps] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor-β (TGFB) regulates cell proliferation, differentiation, apoptosis, and matrix homeostasis and is intimately involved in fibrosis. TGFB expression is increased in fibrotic lung diseases, such as idiopathic pulmonary fibrosis, and in chronic inflammatory conditions, such as chronic obstructive pulmonary disease and asthma. In addition to exhibiting profibrotic activities, the protein exhibits profound immune-suppressive actions involving both innate and adaptive responses, but often this aspect of TGFB biology is overlooked. Recent investigations have demonstrated that TGFB causes wide-ranging immune suppression, including blunting of pivotal early innate IFN responses. These activities permit severe virus infections, often followed by secondary bacterial infections, which may last longer, with augmented inflammation, scarring, fibrosis, and loss of lung function. Strategies to oppose TGFB actions or to enhance IFN responses may help ameliorate the detrimental consequences of infection in patients with diseases characterized by TGFB overexpression, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Belinda J Thomas
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Keiko Kan-O
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Kate L Loveland
- 4 Centre of Reproductive Health, Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| | - Jack A Elias
- 5 Division of Biology and Medicine, Brown University, Providence, Rhode Island
| | - Philip G Bardin
- 1 Monash Lung and Sleep, Monash Medical Centre, Melbourne, Victoria, Australia.,2 Centre for Innate Immunity and Infectious Diseases and.,3 Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia; and
| |
Collapse
|
87
|
Sarıyıldız MA, Yazmalar L, Batmaz İ, Alpaycı M, Burkan YK, Sula B, Kaplan İ, Yıldız M, Akar ZA, Bozkurt M. Serum GDF-15 level in Behçet's disease: relationships between disease activity and clinical parameters. Int J Dermatol 2017; 55:1289-1294. [PMID: 27206990 DOI: 10.1111/ijd.13309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 12/31/2015] [Accepted: 02/03/2016] [Indexed: 11/29/2022]
Abstract
Growth differentiation factor-15 (GDF-15), a member of the transforming growth factor-β superfamily of cytokines, plays an important role in cell growth, signal transduction, and apoptosis regulation. The aim of this study was to evaluate serum GDF-15 levels and their relationships with disease-related variables in patients with Behçet's disease (BD). Forty-six patients diagnosed with BD and 30 demographically matched healthy control subjects participated in the study. GDF-15 levels were measured in blood samples from patients and controls. The Behçet's Disease Current Activity Form (BDCAF) was used to evaluate the disease activity of BD. There were no significant differences between the two groups in C-reactive protein (CRP) level, mean erythrocyte sedimentation rate (ESR), age, body mass index, and mean GDF-15 levels (P > 0.05). Serum GDF-15 levels were positively correlated with findings for peripheral arthritis and CRP, and with BDCAF erythema nodosum, BDCAF arthralgia, and BDCAF arthritis scores. Patients with BD were divided into two groups according to the presence of peripheral arthritis; nine subjects (20%) were positive for peripheral arthritis. Serum ESR, CRP, white blood cell counts, and GDF-15 levels were significantly higher in the group that was positive for peripheral arthritis (P < 0.05). GDF-15 may play a role in the progression and pathway of Behçet's joint involvement and erythema nodosum that is independent of classic inflammatory response measures.
Collapse
Affiliation(s)
| | - Levent Yazmalar
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakır, Turkey
| | - İbrahim Batmaz
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakır, Turkey
| | - Mahmut Alpaycı
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Yüzüncü Yıl University, Van, Turkey
| | - Yahya Kemal Burkan
- Department of Physical Medicine and Rehabilitation, Memorial Hospital, Diyarbakır, Turkey
| | - Bilal Sula
- Department of Dermatology, Dicle University, Diyarbakır, Turkey
| | - İbrahim Kaplan
- Department of Biochemistry, Faculty of Medicine, Dicle University, Diyarbakır, Turkey
| | - Mehmet Yıldız
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakır, Turkey
| | - Zeynel Abidin Akar
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakır, Turkey
| | - Mehtap Bozkurt
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakır, Turkey
| |
Collapse
|
88
|
Abstract
In response to myocardial infarction (MI), time-dependent leukocyte infiltration is critical to program the acute inflammatory response. Post-MI leukocyte density, residence time in the infarcted area, and exit from the infarcted injury predict resolving or nonresolving inflammation. Overactive or unresolved inflammation is the primary determinant in heart failure pathology post-MI. Here, our review describes supporting evidence that the acute inflammatory response also guides the generation of healing and regenerative mediators after cardiac damage. Time-dependent leukocyte density and diversity and the magnitude of myocardial injury is responsible for the resolving and nonresolving pathway in myocardial healing. Post MI, the diversity of leukocytes, such as neutrophils, macrophages, and lymphocytes, has been explored that regulate the clearance of deceased cardiomyocytes by using the classic and reparative pathways. Among the innovative factors and intermediates that have been recognized as essential in acute the self-healing and clearance mechanism, we highlight specialized proresolving mediators as the emerging factor for post-MI reparative mechanisms-translational leukocyte modifiers, such as aging, the source of leukocytes, and the milieu around the leukocytes. In the clinical setting, it is possible that leukocyte diversity is more prominent as a result of risk factors, such as obesity, diabetes, and hypertension. Pharmacologic agents are critical modifiers of leukocyte diversity in healing mechanisms that may impair or stimulate the clearance mechanism. Future research is needed, with a focused approach to understand the molecular targets, cellular effectors, and receptors. A clear understanding of resolving and nonresolving inflammation in myocardial healing will help to develop novel targets with major emphasis on the resolution of inflammation in heart failure pathology.-Tourki, B., Halade, G. Leukocyte diversity in resolving and nonresolving mechanisms of cardiac remodeling.
Collapse
Affiliation(s)
- Bochra Tourki
- Laboratoire des Venins et Biomolécules Thérapeutiques et Plateforme de Physiologie et de Physiopathologie Cardiovasculaires, Institut Pasteur de Tunis, Université Carthage Tunis, Carthage, Tunisia
| | - Ganesh Halade
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
89
|
Nguyen TP, Sieg SF. TGF-β inhibits IL-7-induced proliferation in memory but not naive human CD4 + T cells. J Leukoc Biol 2017; 102:499-506. [PMID: 28588029 DOI: 10.1189/jlb.3a1216-520rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 05/04/2017] [Accepted: 05/06/2017] [Indexed: 02/01/2023] Open
Abstract
TGF-β is a potent suppressor of T cell activation and expansion. Although the antiproliferative effects of TGF-β are well characterized in TCR-activated cells, the effects of TGF-β on T cell proliferation driven by homeostatic cytokines, such as IL-7, are poorly defined. In the current study, we found that TGF-β inhibits IL-7-induced proliferation in memory, but not in naive human CD4+ T cells. TGF-β impaired c-myc induction in all CD4+ T cell maturation subsets, although the impairment was less sustained in naive CD4+ T cells. TGF-β had no discernible effect on IL-7R signaling (p-STAT-5, p-Akt, or p-S6) in memory T cells but selectively enhanced p-S6 signaling in naive T cells. The inhibitory effects of TGF-β on memory T cell proliferation were partially overcome by chemical inhibition of GSK-3, which also led to enhanced c-myc expression. These data suggest that TGF-β could play an important role in limiting homeostatic proliferation of memory T cells. Our observations also point toward a novel strategy to subvert TGF-β-mediated inhibition of memory T cells by targeting GSK-3 for inhibition.
Collapse
Affiliation(s)
- Thao P Nguyen
- Department of Medicine, Department of Pathology, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | - Scott F Sieg
- Department of Medicine, Department of Pathology, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University/University Hospitals Case Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
90
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 377] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
91
|
Zheng Y, Tang L, Mabardi L, Kumari S, Irvine DJ. Enhancing Adoptive Cell Therapy of Cancer through Targeted Delivery of Small-Molecule Immunomodulators to Internalizing or Noninternalizing Receptors. ACS NANO 2017; 11:3089-3100. [PMID: 28231431 PMCID: PMC5647839 DOI: 10.1021/acsnano.7b00078] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Adoptive cell therapy (ACT) has achieved striking efficacy in B-cell leukemias, but less success treating other cancers, in part due to the rapid loss of ACT T-cell effector function in vivo due to immunosuppression in solid tumors. Transforming growth factor-β (TGF-β) signaling is an important mechanism of immune suppression in the tumor microenvironment, but systemic inhibition of TGF-β is toxic. Here we evaluated the potential of targeting a small molecule inhibitor of TGF-β to ACT T-cells using PEGylated immunoliposomes. Liposomes were prepared that released TGF-β inhibitor over ∼3 days in vitro. We compared the impact of targeting these drug-loaded vesicles to T-cells via an internalizing receptor (CD90) or noninternalizing receptor (CD45). When lymphocytes were preloaded with immunoliposomes in vitro prior to adoptive therapy, vesicles targeted to both CD45 and CD90 promoted enhanced T-cell expression of granzymes relative to free systemic drug administration, but only targeting to CD45 enhanced accumulation of granzyme-expressing T-cells in tumors, which correlated with the greatest enhancement of T-cell antitumor activity. By contrast, when administered i.v. to target T-cells in vivo, only targeting of a CD90 isoform expressed exclusively by the donor T-cells led to greater tumor regression over equivalent doses of free systemic drug. These results suggest that in vivo, targeting of receptors uniquely expressed by donor T-cells is of paramount importance for maximal efficacy. This immunoliposome strategy should be broadly applicable to target exogenous or endogenous T-cells and defines parameters to optimize delivery of supporting (or suppressive) drugs to these important immune effectors.
Collapse
Affiliation(s)
- Yiran Zheng
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Li Tang
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Llian Mabardi
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Sudha Kumari
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
| | - Darrell J. Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts, USA
- Department of Material Science and Engineering, MIT, Cambridge, Massachusetts, USA
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard University, Boston, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
- Correspondence should be addressed to D.J.I., Darrell J. Irvine, MIT Room 76-261, 77 Massachusetts Avenue, Cambridge, MA 02139, USA, Telephone: 617-452-4174. Fax: 617-452-3293.
| |
Collapse
|
92
|
Liang XH, Rong L, He G, He H, Lin S, Yang Y, Xue Y, Fang Y. Polymorphisms of the TGF-β1 gene and the risk of acquired aplastic anemia in a Chinese population. Ann Hematol 2017; 96:339-344. [PMID: 27933374 PMCID: PMC5288442 DOI: 10.1007/s00277-016-2886-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/26/2016] [Indexed: 01/19/2023]
Abstract
Acquired aplastic anemia (AA) is a hematological disease characterized by failure of bone marrow hematopoiesis resulting in pancytopenia. While immune-mediated destruction of hematopoietic stem/progenitor cells (HSPCs) plays a central role in the pathophysiology of acquired AA, the transforming growth factor-β1 (TGF-β1) is crucial in adjusting the immune system. The aim of our study was to investigate the role of TGF-β1 gene polymorphisms rs1800469 and rs2317130 in susceptibility to acquired AA. Via the approach of SNaPshot, we genotyped rs1800469 and rs2317130 in 101 patients with acquired AA and 165 controls. It derived us to the conclusion that the genotype TT of rs1800469 (C/T) was significantly associated with decreased risk of acquired AA (adjusted OR = 0.39, 95% CI = 0.18-0.83, P = 0.014). Furthermore, this decreased risk was more pronounced among male patients (adjusted OR = 0.35, 95% CI = 0.13-0.95, P = 0.038) and SAA/vSAA (severe AA/very severe AA) patients (adjusted OR = 0.31, 95% CI = 0.12-0.77, P = 0.02) compared with controls in subgroup analysis. However, a significant increased risk was observed in the genotype distributions of rs2317130 for TT genotype (adjusted OR = 2.52, 95% CI = 1.03-6.19, P = 0.04) compared with the CC genotype among the SAA/vSAA patients and controls in the severity stratification analysis. Our results indicated that TGF-β1 gene polymorphisms might be involved in the munity of acquired AA in a Chinese population. This initial analysis provides valuable clues for further study of TGF-β1 pathway genes in acquired AA.
Collapse
Affiliation(s)
- Xue-Hong Liang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Liucheng Rong
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Guangsheng He
- Department of Hematology, Jiangsu Province Hospital/The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hailong He
- Department of Hematology and Oncology, Soochow Children's Hospital Affiliated to Soochow University, Suzhou, China
| | - Shengyun Lin
- Department of Hematology, First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Yang
- Department of Hematology and Oncology, The First Hospital of Jilin University, Changchun, China
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
93
|
Lee PW, Severin ME, Lovett-Racke AE. TGF-β regulation of encephalitogenic and regulatory T cells in multiple sclerosis. Eur J Immunol 2017; 47:446-453. [PMID: 28102541 DOI: 10.1002/eji.201646716] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 12/01/2016] [Accepted: 01/16/2017] [Indexed: 11/09/2022]
Abstract
Transforming growth factor beta (TGF-β) is a pleiotropic cytokine that has been shown to influence the differentiation and function of T cells. The role that TGF-β plays in immune-mediated disease, such as multiple sclerosis (MS), has become a major area of investigation since CD4+ T cells appear to be a major mediator of autoimmunity. This review provides an analysis of the literature on the role that TGF-β plays in the generation and regulation of encephalitogenic and regulatory T cells (Treg) in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, as well as in T cells of MS patients. Since TGF-β plays a major role in the development and function of both CD4+ effector and Treg, which are defective in MS patients, recent studies have found potential mechanisms to explain the basis for these T-cell defects to establish a foundation for potentially modulating TGF-β signaling to restore normal T-cell function in MS patients.
Collapse
Affiliation(s)
- Priscilla W Lee
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Mary E Severin
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, OH, USA
| |
Collapse
|
94
|
Cantelli G, Crosas-Molist E, Georgouli M, Sanz-Moreno V. TGFΒ-induced transcription in cancer. Semin Cancer Biol 2017; 42:60-69. [PMID: 27586372 PMCID: PMC6137079 DOI: 10.1016/j.semcancer.2016.08.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/19/2016] [Indexed: 12/15/2022]
Abstract
The Transforming Growth Factor-beta (TGFβ) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFβ has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFβ-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFβ target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFβ pathway.
Collapse
Affiliation(s)
- Gaia Cantelli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Eva Crosas-Molist
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Mirella Georgouli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
95
|
O'Connor D, Clutterbuck EA, Thompson AJ, Snape MD, Ramasamy MN, Kelly DF, Pollard AJ. High-dimensional assessment of B-cell responses to quadrivalent meningococcal conjugate and plain polysaccharide vaccine. Genome Med 2017; 9:11. [PMID: 28137280 PMCID: PMC5282650 DOI: 10.1186/s13073-017-0400-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/05/2017] [Indexed: 12/25/2022] Open
Abstract
Background Neisseria meningitidis is a globally important cause of meningitis and septicaemia. Twelve capsular groups of meningococci are known, and quadrivalent vaccines against four of these (A, C, W and Y) are available as plain-polysaccharide and protein-polysaccharide conjugate vaccines. Here we apply contemporary methods to describe B-cell responses to meningococcal polysaccharide and conjugate vaccines. Methods Twenty adults were randomly assigned to receive either a meningococcal plain-polysaccharide or conjugate vaccine; one month later all received the conjugate vaccine. Blood samples were taken pre-vaccination and 7, 21 and 28 days after vaccination; B-cell responses were assessed by ELISpot, serum bactericidal assay, flow cytometry and gene expression microarray. Results Seven days after an initial dose of either vaccine, a gene expression signature characteristic of plasmablasts was detectable. The frequency of newly generated plasma cells (CXCR3+HLA-DR+) and the expression of transcripts derived from IGKC and IGHG2 correlated with immunogenicity. Notably, using an independent dataset, the expression of glucosamine (N-acetyl)-6-sulfatase was found to reproducibly correlate with the magnitude of immune response. Transcriptomic and flow cytometric data revealed depletion of switched memory B cells following plain-polysaccharide vaccine. Conclusions These data describe distinct gene signatures associated with the production of high-avidity antibody and a plain-polysaccharide-specific signature, possibly linked to polysaccharide-induced hyporesponsiveness. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0400-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Daniel O'Connor
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK. .,NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Elizabeth A Clutterbuck
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Amber J Thompson
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Matthew D Snape
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Maheshi N Ramasamy
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Dominic F Kelly
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Churchill Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
96
|
Jung B, Staudacher JJ, Beauchamp D. Transforming Growth Factor β Superfamily Signaling in Development of Colorectal Cancer. Gastroenterology 2017; 152:36-52. [PMID: 27773809 PMCID: PMC5550896 DOI: 10.1053/j.gastro.2016.10.015] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Transforming growth factor (TGF)-β cytokines signal via a complex network of pathways to regulate proliferation, differentiation, adhesion, migration, and other functions in many cell types. A high percentage of colorectal tumors contain mutations that disrupt TGF-β family member signaling. We review how TGF-β family member signaling is altered during development of colorectal cancer, models of study, interaction of pathways, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Barbara Jung
- University of Illinois at Chicago, Chicago, Illinois.
| | | | | |
Collapse
|
97
|
Ogonek J, Kralj Juric M, Ghimire S, Varanasi PR, Holler E, Greinix H, Weissinger E. Immune Reconstitution after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2016; 7:507. [PMID: 27909435 PMCID: PMC5112259 DOI: 10.3389/fimmu.2016.00507] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/02/2016] [Indexed: 12/17/2022] Open
Abstract
The timely reconstitution and regain of function of a donor-derived immune system is of utmost importance for the recovery and long-term survival of patients after allogeneic hematopoietic stem cell transplantation (HSCT). Of note, new developments such as umbilical cord blood or haploidentical grafts were associated with prolonged immunodeficiency due to delayed immune reconstitution, raising the need for better understanding and enhancing the process of immune reconstitution and finding strategies to further optimize these transplant procedures. Immune reconstitution post-HSCT occurs in several phases, innate immunity being the first to regain function. The slow T cell reconstitution is regarded as primarily responsible for deleterious infections with latent viruses or fungi, occurrence of graft-versus-host disease, and relapse. Here we aim to summarize the major steps of the adaptive immune reconstitution and will discuss the importance of immune balance in patients after HSCT.
Collapse
Affiliation(s)
- Justyna Ogonek
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Mateja Kralj Juric
- BMT, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sakhila Ghimire
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | - Pavankumar Reddy Varanasi
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Ernst Holler
- Department of Hematology and Oncology, University of Regensburg, Regensburg, Germany
| | | | - Eva Weissinger
- Transplantation Biology, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
98
|
Doersch KM, Moses KA, Zimmer WE. Synergistic immunologic targets for the treatment of prostate cancer. Exp Biol Med (Maywood) 2016; 241:1900-1910. [PMID: 27444149 PMCID: PMC5068457 DOI: 10.1177/1535370216660212] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Prostate cancer is a common disease and, while detection and treatment have advanced, it remains a significant cause of morbidity and mortality in men. Research suggests significant involvement of the immune system in the pathogenesis and progression of prostate cancer, indicating that immunologic therapies may benefit patients. Two immunologic factors, interleukin-2 and transforming growth factor-β, may be especially attractive therapeutic targets for prostate cancer. Specifically, an increase in interleukin-2 signaling and a decrease in transforming growth factor-β signaling might help improve immunologic recognition and targeting of tumor cells. The purpose of this review is to highlight the evidence that interleukin-2 and blockade of transforming growth factor-β could be used to target prostate cancer based on current understanding of immune function in the context of prostate cancer. Additionally, current treatments related to these two factors for prostate and other cancers will be used to strengthen the argument for this strategy.
Collapse
Affiliation(s)
- Karen M Doersch
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Temple, TX 76504, USA
| | - Kelvin A Moses
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Warren E Zimmer
- Department of Medical Physiology, Texas A&M Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
99
|
Metelli A, Wu BX, Fugle CW, Rachidi S, Sun S, Zhang Y, Wu J, Tomlinson S, Howe PH, Yang Y, Garrett-Mayer E, Liu B, Li Z. Surface Expression of TGFβ Docking Receptor GARP Promotes Oncogenesis and Immune Tolerance in Breast Cancer. Cancer Res 2016; 76:7106-7117. [PMID: 27913437 DOI: 10.1158/0008-5472.can-16-1456] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/23/2016] [Accepted: 09/30/2016] [Indexed: 12/20/2022]
Abstract
GARP encoded by the Lrrc32 gene is the cell surface docking receptor for latent TGFβ, which is expressed naturally by platelets and regulatory T cells (Treg). Although Lrrc32 is amplified frequently in breast cancer, the expression and relevant functions of GARP in cancer have not been explored. Here, we report that GARP exerts oncogenic effects, promoting immune tolerance by enriching and activating latent TGFβ in the tumor microenvironment. We found that human breast, lung, and colon cancers expressed GARP aberrantly. In genetic studies in normal mammary gland epithelial and carcinoma cells, GARP expression increased TGFβ bioactivity and promoted malignant transformation in immunodeficient mice. In breast carcinoma-bearing mice that were immunocompetent, GARP overexpression promoted Foxp3+ Treg activity, which in turn contributed to enhancing cancer progression and metastasis. Notably, administration of a GARP-specific mAb limited metastasis in an orthotopic model of human breast cancer. Overall, these results define the oncogenic effects of the GARP-TGFβ axis in the tumor microenvironment and suggest mechanisms that might be exploited for diagnostic and therapeutic purposes. Cancer Res; 76(24); 7106-17. ©2016 AACR.
Collapse
Affiliation(s)
- Alessandra Metelli
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Bill X Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Caroline W Fugle
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Saleh Rachidi
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Shaoli Sun
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Jennifer Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Yi Yang
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Elizabeth Garrett-Mayer
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Bei Liu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
100
|
Guo H, Lu L, Wang R, Perez-Gutierrez A, Abdulkerim H, Zahorchak A, Sumpter T, Reimann KA, Thomson A, Ezzelarab M. Impact of Human Mutant TGFβ1/Fc Protein on Memory and Regulatory T Cell Homeostasis Following Lymphodepletion in Nonhuman Primates. Am J Transplant 2016; 16:2994-3006. [PMID: 27217298 PMCID: PMC5121100 DOI: 10.1111/ajt.13883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/29/2016] [Accepted: 05/07/2016] [Indexed: 01/25/2023]
Abstract
Transforming growth factor β1 (TGFβ1) plays a key role in T cell homeostasis and peripheral tolerance. We evaluated the influence of a novel human mutant TGFβ1/Fc (human IgG4 Fc) fusion protein on memory CD4+ and CD8+ T cell (Tmem) responses in vitro and their recovery following antithymocyte globulin (ATG)-mediated lymphodepletion in monkeys. TGFβ1/Fc induced Smad2/3 protein phosphorylation in rhesus and human peripheral blood mononuclear cells and augmented the suppressive effect of rapamycin on rhesus Tmem proliferation after either alloactivation or anti-CD3/CD28 stimulation. In combination with IL-2, the incidence of CD4+ CD25hi Foxp3hi regulatory T cells (Treg) and Treg:Th17 ratios were increased. In lymphodepleted monkeys, whole blood trough levels of infused TGFβ1/Fc were maintained between 2 and 7 μg/mL for 35 days. Following ATG administration, total T cell numbers were reduced markedly. In those given TGFβ1/Fc infusion, CD8+ T cell recovery to predepletion levels was delayed compared to controls. Additionally, numbers of CD4+ CD25hi CD127lo Treg increased at 4-6 weeks after depletion but subsequently declined to predepletion levels by 12 weeks. In all monkeys, CD4+ CD25hi Foxp3hi Treg/CD4+ IL-17+ cell ratios were reduced, particularly after stopping TGFβ1/Fc infusion. Thus, human TGFβ1/Fc infusion may delay Tmem recovery following lymphodepletion in nonhuman primates. Combined (low-dose) IL-2 infusion may be required to improve the Treg:Th17 ratio following lymphodepletion.
Collapse
Affiliation(s)
- H. Guo
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - L. Lu
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - R. Wang
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A. Perez-Gutierrez
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - H.S. Abdulkerim
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A.F. Zahorchak
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - T.L. Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine
| | - K. A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - A.W. Thomson
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - M.B. Ezzelarab
- Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA,Corresponding author: Mohamed B. Ezzelarab, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, 200 Lothrop Street, E1558 BST, Pittsburgh, PA 15261,
| |
Collapse
|