1
|
Wu X, Du F, Zhang A, Zhang G, Xu R, Du X. KDELR2 is necessary for chronic obstructive pulmonary disease airway Mucin5AC hypersecretion via an IRE1α/XBP-1s-dependent mechanism. J Cell Mol Med 2024; 28:e70125. [PMID: 39365189 PMCID: PMC11451269 DOI: 10.1111/jcmm.70125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
Airway mucus hypersecretion, a crucial pathological feature of chronic obstructive pulmonary disease (COPD), contributes to the initiation, progression, and exacerbation of this disease. As a macromolecular mucin, the secretory behaviour of Mucin5AC (MUC5AC) is highly dependent on a series of modifying and folding processes that occur in the endoplasmic reticulum (ER). In this study, we focused on the ER quality control protein KDEL receptor (KDELR) and demonstrated that KDELR2 and MUC5AC were colocalized in the airway epithelium of COPD patients and COPD model rats. In addition, knockdown of KDELR2 markedly reduced the expression of MUC5AC both in vivo and in vitro and knockdown of ATF6 further decreased the levels of KDELR2. Furthermore, pretreatment with 4μ8C, an IRE1α inhibitor, led to a partial reduction in the expression of KDELR2 and MUC5AC both in vivo and in vitro, which indicated the involvement of IRE1α/XBP-1s in the upstream signalling cascade. Our study revealed that KDELR2 plays a crucial role in airway MUC5AC hypersecretion in COPD, which might be dependent on ATF6 and IRE1α/XBP-1s upstream signalling.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Fawang Du
- Department of Respiratory and Critical Care MedicineSuining Central HospitalSuiningSichuanChina
| | - Aijie Zhang
- Basic Laboratory, Key Laboratory of Metabolic DiseasesSuining Central HospitalSuiningChina
| | - Guoyue Zhang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Rui Xu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xianzhi Du
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
2
|
Choi HJ, Wu Y, McDaniel Mims B, Pugel A, Tang CHA, Tian L, Hu CCA, Yu XZ. Endoplasmic Reticulum Stress Response Mediator IRE-1α Promotes Host Dendritic Cells in Graft-versus-Host Disease Development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:384-393. [PMID: 38864663 PMCID: PMC11415232 DOI: 10.4049/jimmunol.2300616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 05/22/2024] [Indexed: 06/13/2024]
Abstract
Allogeneic hematopoietic cell transplantation is an effective treatment for hematologic malignancies, but the complications such as graft-versus-host disease (GVHD) can limit its benefit. The conditioning regimens before transplant, including chemotherapy or irradiation, can trigger endoplasmic reticulum stress. IRE-1α is a major endoplasmic reticulum stress mediator that can further activate both spliced XBP-1 (XBP-1s) and regulated IRE-1-dependent decay (RIDD). IRE-1α-XBP-1s signaling controls dendritic cell (DC) differentiation and Ag presentation, crucial in GVHD progression. In this study, we used DC-specific XBP-1-deficient mice as donors or recipients and observed that XBP-1s was crucial for host DCs in the induction of GVHD but dispensable for the graft-versus-leukemia response. To specifically target IRE-1α in the host, we treated recipient mice with the IRE-1α inhibitor B-I09 for 3 d prior to bone marrow transplantation, which significantly suppressed GVHD development while maintaining the graft-versus-leukemia effect. XBP-1-deficient or BI09-treated recipients showed reduced DC survival after irradiation and bone marrow transplantation. Inhibition of IRE-1α also led to a reduction in DC alloreactivity, subsequently decreasing the proliferation and activation of allogeneic T cells. With further study using RIDD-deficient DCs, we observed that RIDD was also required for optimal DC activation. Taken together, XBP-1s and RIDD both promote host DC survival and alloreactivity that contribute to GVHD development.
Collapse
Affiliation(s)
- Hee-Jin Choi
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Brianyell McDaniel Mims
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC
| | - Allison Pugel
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Chih-Hang Anthony Tang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX
| | - Linlu Tian
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Chih-Chi Andrew Hu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Neal Cancer Center, Houston Methodist Research Institute, Houston, TX
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
3
|
Choi HJ, Yu XZ. ER stress: an emerging regulator in GVHD development. Front Immunol 2023; 14:1212215. [PMID: 37744326 PMCID: PMC10511645 DOI: 10.3389/fimmu.2023.1212215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is a promising therapeutic option for hematologic malignancies. However, the clinical benefits of allo-HCT are limited by the development of complications including graft-versus-host disease (GVHD). Conditioning regimens, such as chemotherapy and irradiation, which are administered to the patients prior to allo-HCT, can disrupt the endoplasmic reticulum (ER) homeostasis, and induce ER stress in the recipient's cells. The conditioning regimen activates antigen-presenting cells (APCs), which, in turn, activate donor cells, leading to ER stress in the transplanted cells. The unfolded protein response (UPR) is an evolutionarily conserved signaling pathway that manages ER stress in response to cellular stress. UPR has been identified as a significant regulatory player that influences the function of various immune cells, including T cells, B cells, macrophages, and dendritic cells (DCs), in various disease progressions. Therefore, targeting the UPR pathway has garnered significant attention as a promising approach for the treatment of numerous diseases, such as cancer, neurodegeneration, diabetes, and inflammatory diseases. In this review, we summarize the current literature regarding the contribution of ER stress response to the development of GVHD in both hematopoietic and non-hematopoietic cells. Additionally, we explore the potential therapeutic implications of targeting UPR to enhance the effectiveness of allo-HCT for patients with hematopoietic malignancies.
Collapse
Affiliation(s)
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Department of Medicine, and the Cancer Center, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
4
|
Wu Y, Mealer C, Schutt S, Wilson CL, Bastian D, Sofi MH, Zhang M, Luo Z, Choi HJ, Yang K, Tian L, Nguyen H, Helke K, Schnapp LM, Wang H, Yu XZ. MicroRNA-31 regulates T-cell metabolism via HIF1α and promotes chronic GVHD pathogenesis in mice. Blood Adv 2022; 6:3036-3052. [PMID: 35073581 PMCID: PMC9131913 DOI: 10.1182/bloodadvances.2021005103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 01/10/2022] [Indexed: 11/20/2022] Open
Abstract
Chronic graft-versus-host disease (cGVHD) remains a major obstacle impeding successful allogeneic hematopoietic cell transplantation (HCT). MicroRNAs (miRs) play key roles in immune regulation during acute GVHD development. Preclinical studies to identify miRs that affect cGVHD pathogenesis are required to develop these as potential lifesaving interventions. Using oligonucleotide array, we identified miR-31, which was significantly elevated in allogeneic T cells after HCT in mice. Using genetic and pharmacologic approaches, we demonstrated a key role for miR-31 in mediating donor T-cell pathogenicity in cGVHD. Recipients of miR-31-deficient T cells displayed improved cutaneous and pulmonary cGVHD. Deficiency of miR-31 reduced T-cell expansion and T helper 17 (Th17) cell differentiation but increased generation and function of regulatory T cells (Tregs). MiR-31 facilitated neuropilin-1 downregulation, Foxp3 loss, and interferon-γ production in alloantigen-induced Tregs. Mechanistically, miR-31 was required for hypoxia-inducible factor 1α (HIF1α) upregulation in allogeneic T cells. Therefore, miR-31-deficient CD4 T cells displayed impaired activation, survival, Th17 cell differentiation, and glycolytic metabolism under hypoxia. Upregulation of factor-inhibiting HIF1, a direct target of miR-31, in miR-31-deficient T cells was essential for attenuating T-cell pathogenicity. However, miR-31-deficient CD8 T cells maintained intact glucose metabolism, cytolytic activity, and graft-versus-leukemia response. Importantly, systemic administration of a specific inhibitor of miR-31 effectively reduced donor T-cell expansion, improved Treg generation, and attenuated cGVHD. Taken together, miR-31 is a key driver for T-cell pathogenicity in cGVHD but not for antileukemia activity. MiR-31 is essential in driving cGVHD pathogenesis and represents a novel potential therapeutic target for controlling cGVHD.
Collapse
Affiliation(s)
- Yongxia Wu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Corey Mealer
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - Steven Schutt
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | | | - David Bastian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - M. Hanief Sofi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - Mengmeng Zhang
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - Zhenwu Luo
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - Hee-Jin Choi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Kaipo Yang
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Linlu Tian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Hung Nguyen
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
| | - Kris Helke
- Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC
| | | | - Honglin Wang
- Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC; and
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
5
|
Ceramide synthase 6 impacts T-cell allogeneic response and graft-versus-host disease through regulating N-RAS/ERK pathway. Leukemia 2022; 36:1907-1915. [PMID: 35513703 PMCID: PMC9256768 DOI: 10.1038/s41375-022-01581-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/09/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective immunotherapy for various hematologic malignances, predominantly through potent graft-versus-leukemia (GVL) effect. However, the mortality after allo-HCT is because of relapse of primary malignancy and followed by graft-vs-host-disease (GVHD) as a major cause of transplant-related mortality. Hence, strategies to limit GVHD while preserving the GVL effect are highly desirable. Ceramide, which serves a central role in sphingolipid metabolism, is generated by ceramide synthases (CerS1–6). In this study, we found that genetic or pharmacologic targeting of CerS6 prevented and reversed chronic GVHD (cGVHD). Furthermore, specific inhibition of CerS6 with ST1072 significantly ameliorated acute GVHD (aGVHD) while preserving the GVL effect, which differed from FTY720 that attenuated aGVHD but impaired GVL activity. At the cellular level, blockade of CerS6 restrained donor T cells from migrating into GVHD target organs and preferentially reduced activation of donor CD4 T cells. At the molecular level, CerS6 was required for optimal TCR signaling, CD3/PKCθ co-localization, and subsequent N-RAS activation and ERK signaling, especially on CD4+ T cells. The current study provides rationale and means for targeting CerS6 to control GVHD and leukemia relapse, which would enhance the efficacy of allo-HCT as an immunotherapy for hematologic malignancies in the clinic.
Collapse
|
6
|
Shao A, Xu Q, Kang CW, Cain CF, Lee AC, Tang CHA, Del Valle JR, Hu CCA. IRE-1-Targeting Caged Prodrug with Endoplasmic Reticulum Stress-Inducing and XBP-1S-Inhibiting Activities for Cancer Therapy. Mol Pharm 2022; 19:1059-1067. [PMID: 35253431 PMCID: PMC9296017 DOI: 10.1021/acs.molpharmaceut.1c00639] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Activation
of the IRE-1/XBP-1s pathway supports tumor progression.
Here, we report a novel prodrug, TC-D-F07, in which a thiol-reactive
dinitrobenzenesulfonyl (Dns) cage was installed onto the C8 hydroxyl
of the covalent IRE-1 inhibitor D-F07. The electron-withdrawing Dns
group in TC-D-F07 stabilizes the neighboring 1,3-dioxane acetal, allowing
for stimulus-mediated control of its inhibitory activity. TC-D-F07
exhibits high sensitivity to intracellular thiols. Because tumor cells
exhibit higher concentrations of glutathione and cysteine, treatment
with TC-D-F07 results in more sustained levels of D-F07 in transformed
versus normal cells. In addition, we show that a dinitrophenyl cysteine
adduct resulting from cleavage of the Dns group induces endoplasmic
reticulum (ER) stress, causing tumor cells to increase the expression
of XBP-1s. The accumulated levels of D-F07 and its gradual decomposition
into the active IRE-1 inhibitor eventually deprive tumor cells of
XBP-1s, leading to more severe apoptosis than those treated with its
uncaged analogue.
Collapse
Affiliation(s)
- Andong Shao
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Qin Xu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Chang Won Kang
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Christopher F. Cain
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Avery C. Lee
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Chih-Hang Anthony Tang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Juan R. Del Valle
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Chih-Chi Andrew Hu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| |
Collapse
|
7
|
Choi HJ, Tang CHA, Tian L, Wu Y, Sofi MH, Ticer T, Schutt SD, Hu CCA, Yu XZ. XBP-1s Promotes B Cell Pathogenicity in Chronic GVHD by Restraining the Activity of Regulated IRE-1α-Dependent Decay. Front Immunol 2021; 12:705484. [PMID: 34659198 PMCID: PMC8517405 DOI: 10.3389/fimmu.2021.705484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/15/2021] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is an effective therapeutic procedure to treat hematological malignancies. However, the benefit of allo-HCT is limited by a major complication, chronic graft-versus-host disease (cGVHD). Since transmembrane and secretory proteins are generated and modified in the endoplasmic reticulum (ER), the ER stress response is of great importance to secretory cells including B cells. By using conditional knock-out (KO) of XBP-1, IRE-1α or both specifically on B cells, we demonstrated that the IRE-1α/XBP-1 pathway, one of the major ER stress response mediators, plays a critical role in B cell pathogenicity on the induction of cGVHD in murine models of allo-HCT. Endoribonuclease activity of IRE-1α activates XBP-1 signaling by converting unspliced XBP-1 (XBP-1u) mRNA into spliced XBP-1 (XBP-1s) mRNA but also cleaves other ER-associated mRNAs through regulated IRE-1α-dependent decay (RIDD). Further, ablation of XBP-1s production leads to unleashed activation of RIDD. Therefore, we hypothesized that RIDD plays an important role in B cells during cGVHD development. In this study, we found that the reduced pathogenicity of XBP-1 deficient B cells in cGVHD was reversed by RIDD restriction in IRE-1α kinase domain KO mice. Restraining RIDD activity per se in B cells resulted in an increased severity of cGVHD. Besides, inhibition of RIDD activity compromised B cell differentiation and led to dysregulated expression of MHC II and costimulatory molecules such as CD86, CD40, and ICOSL in B cells. Furthermore, restraining the RIDD activity without affecting XBP-1 splicing increased B cell ability to induce cGVHD after allo-HCT. These results suggest that RIDD is an important mediator for reducing cGVHD pathogenesis through targeting XBP-1s.
Collapse
Affiliation(s)
- Hee-Jin Choi
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Chih-Hang Anthony Tang
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Linlu Tian
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Yongxia Wu
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - M Hanief Sofi
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Taylor Ticer
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Steven D Schutt
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Chih-Chi Andrew Hu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, United States
| | - Xue-Zhong Yu
- Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, United States.,Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
8
|
BAFF promotes heightened BCR responsiveness and manifestations of chronic GVHD after allogeneic stem cell transplantation. Blood 2021; 137:2544-2557. [PMID: 33534893 PMCID: PMC8109011 DOI: 10.1182/blood.2020008040] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/21/2021] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic graft-versus-host disease (cGVHD) have increased B cell-activating factor (BAFF) levels, but whether BAFF promotes disease after allogeneic bone marrow transplantation (allo-BMT) remains unknown. In a major histocompatibility complex-mismatched model with cGVHD-like manifestations, we first examined B-lymphopenic μMT allo-BMT recipients and found that increased BAFF levels in cGVHD mice were not merely a reflection of B-cell number. Mice that later developed cGVHD had significantly increased numbers of recipient fibroblastic reticular cells with higher BAFF transcript levels. Increased BAFF production by donor cells also likely contributed to cGVHD, because BAFF transcript in CD4+ T cells from diseased mice and patients was increased. cGVHD manifestations in mice were associated with high BAFF/B-cell ratios and persistence of B-cell receptor (BCR)-activated B cells in peripheral blood and lesional tissue. By employing BAFF transgenic (Tg) mice donor cells, we addressed whether high BAFF contributed to BCR activation in cGVHD. BAFF increased NOTCH2 expression on B cells, augmenting BCR responsiveness to surrogate antigen and NOTCH ligand. BAFF Tg B cells had significantly increased protein levels of the proximal BCR signaling molecule SYK, and high SYK protein was maintained by BAFF after in vitro BCR activation or when alloantigen was present in vivo. Using T cell-depleted (BM only) BAFF Tg donors, we found that BAFF promoted cGVHD manifestations, circulating GL7+ B cells, and alloantibody production. We demonstrate that pathologic production of BAFF promotes an altered B-cell compartment and augments BCR responsiveness. Our findings compel studies of therapeutic targeting of BAFF and BCR pathways in patients with cGVHD.
Collapse
|
9
|
Activation of TREM-1 induces endoplasmic reticulum stress through IRE-1α/XBP-1s pathway in murine macrophages. Mol Immunol 2021; 135:294-303. [PMID: 33957479 DOI: 10.1016/j.molimm.2021.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/16/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Increasing evidence suggests that endoplasmic reticulum (ER) stress activates several pro-inflammatory signaling pathways in many diseases, including acute lung injury (ALI). We have reported that blocking triggering receptor expressed on myeloid cells 1 (TREM-1) protects against ALI by suppressing pulmonary inflammation in mice with ALI induced by lipopolysaccharides (LPS). However, the molecular mechanism underlying the TREM-1-induced pro-inflammatory microenvironment in macrophages remains unclearly. Herein, we aimed to determine whether TREM-1 regulates the inflammatory responses induced by LPS associated with ER stress activation. We found that the activation of TREM-1 by a monoclonal agonist antibody (anti-TREM-1) increased the mRNA and protein levels of IL-1β, TNF-α, and IL-6 in primary macrophages. Treatment of the anti-TREM-1 antibody increased the expression of ER stress markers (ATF6, PERK, IRE-1α, and XBP-1s) in primary macrophages. While pretreatment with 4-PBA, an inhibitor of ER stress, significantly inhibited the expression of ER stress markers and pro-inflammatory cytokines and reduced LDH release. Furthermore, inhibiting the activity of the IRE-1α/XBP-1s pathway by STF-083010 significantly mitigated the increased levels of IL-1β, TNF-α, and IL-6 in macrophages treated by the anti-TREM-1 antibody. XBP-1 silencing attenuated pro-inflammatory microenvironment evoked by activation of TREM-1. Besides, we found that blockade of TREM-1 with LR12 ameliorated ER stress induced by LPS in vitro and in vivo. In conclusion, we conclude that TREM-1 activation induces ER stress through the IRE-1α/XBP-1s pathway in macrophages, contributing to the pro-inflammatory microenvironment.
Collapse
|
10
|
Del Valle JR, Betts BC, Yu XZ, Janssens S, Lambrecht BN, Simon MC, Hu CCA. Clarifying the translational potential of B-I09. Nat Chem Biol 2020; 16:1152. [PMID: 33067597 DOI: 10.1038/s41589-020-00654-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Juan R Del Valle
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Brian C Betts
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | | | | | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
11
|
Gardner A, de Mingo Pulido Á, Ruffell B. Dendritic Cells and Their Role in Immunotherapy. Front Immunol 2020; 11:924. [PMID: 32508825 PMCID: PMC7253577 DOI: 10.3389/fimmu.2020.00924] [Citation(s) in RCA: 274] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Despite significant advances in the field of cancer immunotherapy, the majority of patients still do not benefit from treatment and must rely on traditional therapies. Dendritic cells have long been a focus of cancer immunotherapy due to their role in inducing protective adaptive immunity, but cancer vaccines have shown limited efficacy in the past. With the advent of immune checkpoint blockade and the ability to identify patient-specific neoantigens, new vaccines, and combinatorial therapies are being evaluated in the clinic. Dendritic cells are also emerging as critical regulators of the immune response within tumors. Understanding how to augment the function of these intratumoral dendritic cells could offer new approaches to enhance immunotherapy, in addition to improving the cytotoxic and targeted therapies that are partially dependent upon a robust immune response for their efficacy. Here we will discuss the role of specific dendritic cell subsets in regulating the anti-tumor immune response, as well as the current status of dendritic cell-based immunotherapies, in order to provide an overview for future lines of research and clinical trials.
Collapse
Affiliation(s)
- Alycia Gardner
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.,Cancer Biology PhD Program, University of South Florida, Tampa, FL, United States
| | - Álvaro de Mingo Pulido
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.,Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
12
|
Iamsawat S, Tian L, Daenthanasanmak A, Wu Y, Nguyen HD, Bastian D, Yu XZ. Vitamin C stabilizes CD8+ iTregs and enhances their therapeutic potential in controlling murine GVHD and leukemia relapse. Blood Adv 2019; 3:4187-4201. [PMID: 31856270 PMCID: PMC6929397 DOI: 10.1182/bloodadvances.2019000531] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Adoptive transfer of induced regulatory T cells (iTregs) can ameliorate graft-versus-host disease (GVHD) after allogeneic hematopoietic cell transplantation (allo-HCT). CD4+ iTregs can effectively prevent GVHD but impair the graft-versus-leukemia (GVL) effect, whereas CD8+ iTregs preserve the GVL effect but have limited efficacy in GVHD control because of their instability under inflammatory conditions. Thus, we aimed to stabilize CD8+ iTregs via treatment with vitamin C (Vit C) to improve their efficacy in controlling GVHD. We found that addition of Vit C significantly improved the stability of forkhead box P3 (Foxp3) expression in CD8+ iTregs. Moreover, Vit C-treated CD8+ iTregs exhibited high efficacy in attenuating acute and chronic GVHD. The mechanistic study revealed that addition of Vit C to CD8+ iTreg culture markedly increased DNA demethylation in the conserved noncoding sequence 2 region and, hence, maintained higher Foxp3 expression levels compared with untreated controls. In acute GVHD, Vit C-treated CD8+ iTregs were able to inhibit pathogenic T-cell expansion and differentiation while reducing thymus damage and B-cell activation in cGVHD. Importantly, in contrast to CD4+ iTregs, Vit C-treated CD8+ iTregs retained the ability to control tumor relapse. These results provide a strong rationale to use Vit C in the clinic to stabilize CD8+ iTregs for the control of GVHD and preservation of GVL after allo-HCT.
Collapse
Affiliation(s)
| | - Linlu Tian
- Department of Microbiology and Immunology and
| | | | - Yongxia Wu
- Department of Microbiology and Immunology and
| | | | | | - Xue-Zhong Yu
- Department of Microbiology and Immunology and
- Department of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
13
|
Park K, Lee SE, Shin KO, Uchida Y. Insights into the role of endoplasmic reticulum stress in skin function and associated diseases. FEBS J 2019; 286:413-425. [PMID: 30586218 DOI: 10.1111/febs.14739] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/29/2018] [Accepted: 12/19/2018] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum (ER) stress is a mechanism that allows the protection of normal cellular functions in response to both internal perturbations, such as accumulation of unfolded proteins, and external perturbations, for example redox stress, UVB irradiation, and infection. A hallmark of ER stress is the accumulation of misfolded and unfolded proteins. Physiological levels of ER stress trigger the unfolded protein response (UPR) that is required to restore normal ER functions. However, the UPR can also initiate a cell death program/apoptosis pathway in response to excessive or persistent ER stress. Recently, it has become evident that chronic ER stress occurs in several diseases, including skin diseases such as Darier's disease, rosacea, vitiligo and melanoma; furthermore, it is suggested that ER stress is directly involved in the pathogenesis of these disorders. Here, we review the role of ER stress in skin function, and discuss its significance in skin diseases.
Collapse
Affiliation(s)
- Kyungho Park
- Department of Food Science and Nutrition, Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Sang Eun Lee
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Yoshikazu Uchida
- Department of Dermatology, School of Medicine, University of California, San Francisco, CA, USA.,Northern California Institute for Research and Education, Veterans Affairs Medical Center, San Francisco, CA, USA
| |
Collapse
|
14
|
Shao A, Kang CW, Tang CHA, Cain CF, Xu Q, Phoumyvong CM, Del Valle JR, Hu CCA. Structural Tailoring of a Novel Fluorescent IRE-1 RNase Inhibitor to Precisely Control Its Activity. J Med Chem 2019; 62:5404-5413. [PMID: 31083990 DOI: 10.1021/acs.jmedchem.9b00269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Activation of the IRE-1/XBP-1 pathway has been linked to many human diseases. We report a novel fluorescent tricyclic chromenone inhibitor, D-F07, in which we incorporated a 9-methoxy group onto the chromenone core to enhance its potency and masked the aldehyde to achieve long-term efficacy. Protection of the aldehyde as a 1,3-dioxane acetal led to strong fluorescence emitted by the coumarin chromophore, enabling D-F07 to be tracked inside the cell. We installed a photolabile structural cage on the hydroxy group of D-F07 to generate PC-D-F07. Such a modification significantly stabilized the 1,3-dioxane acetal protecting group, allowing for specific stimulus-mediated control of inhibitory activity. Upon photoactivation, the re-exposed hydroxy group on D-F07 triggered the aldehyde-protecting 1,3-dioxane acetal to slowly decompose, leading to the inhibition of the RNase activity of IRE-1. Our novel findings will also allow for spatiotemporal control of the inhibitory effect of other salicylaldehyde-based compounds currently in development.
Collapse
Affiliation(s)
- Andong Shao
- The Wistar Institute , 3601 Spruce Street , Philadelphia , Pennsylvania 19104 , United States
| | - Chang Won Kang
- Department of Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Chih-Hang Anthony Tang
- The Wistar Institute , 3601 Spruce Street , Philadelphia , Pennsylvania 19104 , United States
| | - Christopher F Cain
- Department of Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Qin Xu
- The Wistar Institute , 3601 Spruce Street , Philadelphia , Pennsylvania 19104 , United States
| | - Claire M Phoumyvong
- The Wistar Institute , 3601 Spruce Street , Philadelphia , Pennsylvania 19104 , United States
| | - Juan R Del Valle
- Department of Chemistry , University of South Florida , Tampa , Florida 33620 , United States
| | - Chih-Chi Andrew Hu
- The Wistar Institute , 3601 Spruce Street , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
15
|
Sofi MH, Wu Y, Schutt SD, Dai M, Daenthanasanmak A, Heinrichs Voss J, Nguyen H, Bastian D, Iamsawat S, Selvam SP, Liu C, Maulik N, Ogretmen B, Jin J, Mehrotra S, Yu XZ. Thioredoxin-1 confines T cell alloresponse and pathogenicity in graft-versus-host disease. J Clin Invest 2019; 129:2760-2774. [PMID: 31045571 DOI: 10.1172/jci122899] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress is elevated in the recipients of allogeneic hematopoietic transplantation (allo-HCT) and likely contributes to the development of graft-versus-host disease (GVHD). GVHD is characterized by activation, expansion, cytokine production and migration of alloreactive donor T cells, and remains a major cause of morbidity and mortality after allo-HCT. Hence, strategies to limit oxidative stress in GVHD are highly desirable. Thioredoxin1 (Trx1) counteracts oxidative stress by scavenging reactive oxygen species (ROS) and regulating other enzymes that metabolize H2O2. The present study sought to elucidate the role of Trx1 in the pathophysiology of GVHD. Using murine and xenograft models of allogeneic bone marrow transplantation (allo-BMT) and genetic (human Trx1-transgenic, Trx1-Tg) as well as pharmacologic (human recombinant Trx1, RTrx1) strategies; we found that Trx1-Tg donor T cells or administration of the recipients with RTrx1 significantly reduced GVHD severity. Mechanistically, we observed RTrx1 reduced ROS accumulation and cytokine production of mouse and human T cells in response to alloantigen stimulation in vitro. In allo-BMT settings, we found that Trx1-Tg or RTrx1 decreased downstream signaling molecules including NFκB activation and T-bet expression, and reduced proliferation, IFN-γ production and ROS accumulation in donor T cells within GVHD target organs. More importantly, administration of RTrx1 did not impair the graft-versus-leukemia (GVL) effect. Taken together, the current work provides a strong rationale and demonstrates feasibility to target the ROS pathway, which can be readily translated into clinic.
Collapse
Affiliation(s)
| | - Yongxia Wu
- Department of Microbiology and Immunology and
| | | | - Min Dai
- Department of Microbiology and Immunology and
| | | | | | - Hung Nguyen
- Department of Microbiology and Immunology and
| | | | | | - Shanmugam Panneer Selvam
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School and Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Nilanjana Maulik
- Department of Surgery, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | | | - Xue-Zhong Yu
- Department of Microbiology and Immunology and.,Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
16
|
Daenthanasanmak A, Iamsawat S, Chakraborty P, Nguyen HD, Bastian D, Liu C, Mehrotra S, Yu XZ. Targeting Sirt-1 controls GVHD by inhibiting T-cell allo-response and promoting Treg stability in mice. Blood 2019; 133:266-279. [PMID: 30514750 PMCID: PMC6337874 DOI: 10.1182/blood-2018-07-863233] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Graft-versus-host disease (GVHD) remains one of the major complications after allogeneic bone marrow transplantation (allo-BMT). Sirtuin-1 (Sirt-1) plays a crucial role in various biological processes including cellular senescence, metabolism, and inflammatory responses. Sirt-1 deacetylation regulates different transcription factors that are important for modulating immune responses. In the current study, we addressed the role of Sirt-1 in GVHD induction by employing Sirt-1 conditional knockout mice as well as a pharmacological Sirt-1 inhibitor. Using major histocompatibility complex (MHC)-mismatched and MHC-matched murine BMT models, we found that Sirt-1-/- T cells had a reduced ability to induce acute GVHD (aGVHD) via enhanced p53 acetylation. Sirt-1-deficient T cells also promoted induced regulatory T cell (iTreg) differentiation and inhibited interferon-γ production after allo-BMT. Sirt-1 deletion in iTregs increased Foxp3 stability and restrained iTreg conversion into pathogenic T cells. Furthermore, we found that administration with a Sirt-1 inhibitor, Ex-527, significantly improved recipient survival and clinical scores, with no signs of tumor relapse. These results indicate that Sirt-1 inhibition can attenuate GVHD while preserving the graft-versus-leukemia effect. Consistently, Sirt-1-deficient T cells also displayed a remarkably reduced ability to induce chronic GVHD (cGVHD). Mechanistic studies revealed that Sirt-1 deficiency in T cells enhanced splenic B-cell reconstitution and reduced follicular T helper cell development. Sirt-1 deficiency in T cells modulated donor B-cell responses reducing both B-cell activation and plasma cell differentiation. In addition, therapeutic Sirt-1 inhibition could both prevent cGVHD and reduce established cGVHD. In conclusion, Sirt-1 is a promising therapeutic target for the control of aGVHD and cGVHD pathogenesis and possesses high potential for clinical application.
Collapse
Affiliation(s)
| | | | - Paramita Chakraborty
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | | | | | - Chen Liu
- Department of Pathology and Laboratory Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ; and
| | - Shikhar Mehrotra
- Department of Surgery, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology and
- Department of Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
17
|
Chang YJ, Zhao XY, Huang XJ. Strategies for Enhancing and Preserving Anti-leukemia Effects Without Aggravating Graft-Versus-Host Disease. Front Immunol 2018; 9:3041. [PMID: 30619371 PMCID: PMC6308132 DOI: 10.3389/fimmu.2018.03041] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curable method for the treatment of hematological malignancies. In the past two decades, the establishment of haploidentical transplant modalities make “everyone has a donor” become a reality. However, graft-versus-host disease (GVHD) and relapse remain the major two causes of death either in the human leukocyte antigen (HLA)-matched transplant or haploidentical transplant settings, both of which restrict the improvement of transplant outcomes. Preclinical mice model showed that both donor-derived T cells and natural killer (NK) cells play important role in the pathogenesis of GVHD and the effects of graft-versus-leukemia (GVL). Hence, understanding the immune mechanisms of GVHD and GVL would provide potential strategies for the control of leukemia relapse without aggravating GVHD. The purpose of the current review is to summarize the biology of GVHD and GVL responses in preclinical models and to discuss potential novel therapeutic strategies to reduce the relapse rate after allo-SCT. We will also review the approaches, including optimal donor selection and, conditioning regimens, donor lymphocyte infusion, BCR/ABL-specific CTL, and chimeric antigen receptor-modified T cells, which have been successfully used in the clinic to enhance and preserve anti-leukemia activity, especially GVL effects, without aggravating GVHD or alleviate GVHD.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
18
|
Betts BC, Locke FL, Sagatys EM, Pidala J, Walton K, Menges M, Reff J, Saha A, Djeu JY, Kiluk JV, Lee MC, Kim J, Kang CW, Tang CH, Frieling J, Lynch CC, List A, Rodriguez PC, Blazar BR, Conejo-Garcia JR, Del Valle JR, Hu CC, Anasetti C. Inhibition of Human Dendritic Cell ER Stress Response Reduces T Cell Alloreactivity Yet Spares Donor Anti-tumor Immunity. Front Immunol 2018; 9:2887. [PMID: 30574153 PMCID: PMC6291501 DOI: 10.3389/fimmu.2018.02887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1β, TGFβ, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.
Collapse
Affiliation(s)
- Brian C Betts
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Elizabeth M Sagatys
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, United States
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Kelly Walton
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Meghan Menges
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Julie Y Djeu
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - John V Kiluk
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Marie C Lee
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - Chang Won Kang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Hang Tang
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Jeremy Frieling
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Conor C Lynch
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Alan List
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, United States
| | - Paulo C Rodriguez
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | | | - Juan R Del Valle
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Chi Hu
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|