Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cell Reports, Elsevier BV, Vol. 40, No. 7 ( 2022-08), p. 111201-
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2649101-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A982-A982
    Abstract: Stimulatory dendritic cells (SDC), enriched within the Batf3-DC lineage (also known as conventional type 1 DC, cDC1), engage in productive interactions with CD8+ effectors along tumor-stroma boundaries. This puzzling pattern of T-cell-DC localization has been interpreted as ”tumor-exclusion”, limiting anti-tumor immunity. To understand this paradox, we hypothesized that dynamic matrix remodeling at the invasive margin generates unique activation and cell-fate cues critical for Batf3-DC homeostasis. Methods We studied immunocompetent tumor models of lung carcinoma, breast carcinoma, melanoma and multiple myeloma. For mechanistic experiments, we generated novel Vcan-targeted models through CRISPR-Cas9 targeting. We delineated DC subsets through multi-parametric flow cytometry and tumor immune contexture through mass cytometry. Batf3-DC cellular models included MutuDC1940 immortalized DC and iCD103 primary cells. TCGA data were mined for human validation. Results We find that CD8+ T cells massively infiltrate tumor matrices undergoing robust matrix proteoglycan versican (VCAN) proteolysis, an essential organ-sculpting modification in development and adult tissue-plane forging. Across 7591 samples from 20 TCGA cancer types, a significant-positive correlation between VCAN substrate expression and Batf3-DC score was observed, suggesting that the VCAN pathway may regulate Batf3-DC across several cancer types. Experimental Vcan depletion in the tumor microenvironment was detrimental for Batf3-DC. Batf3-DC abundance was restored through the VCAN N-terminal fragment (matrikine) versikine, physiologically generated through ADAMTS protease activity in remodeled stroma. In addition to Batf3-DC expansion, versikine resulted in G-MDSC contraction as well as the emergence of an atypical innate lymphoid (NK/ILC1) subset expressing cytotoxicity receptors, low IFNgamma and robust pro-survival GM-CSF. Despite broad intratumoral IRF8 induction (10-100-fold), adoptive transfer of pre-DC into versikine-replete microenvironments did not influence their differentiation choice between Batf3-DC and cDC2. Instead, versikine delivered a distinct Batf3-DC activation signal characterized by non-TLR maturation as well as downregulation of TGFbeta and Wnt signaling. In vivo, versikine promoted Batf3-DC abundance through NK cells but independently of stromal TLR2 or CD44. Versikine sensitized immune-evasive tumors to STING agonist immunotherapy in a Batf3-DC dependent manner and promoted antigen-specific CD8+ responses. Versikine-DC signatures correlated with CD8+ T cell scores in human lung cancers. Conclusions We demonstrate that dynamic extracellular matrix remodeling controls Batf3-DC abundance in the tumor microenvironment. N-terminal proteolysis of the matrix proteoglycan versican (VCAN), releases a bioactive fragment (matrikine), versikine, that is remarkably necessary and sufficient for Batf3-DC accumulation. Versikine orchestrates a multi-lineage network that regulates Batf3-DC activation and survival at matrix-remodeling interfaces. Therapeutic harnessing of matrix-Batf3-DC cross-talk sensitizes immune-evasive tumors to immunotherapy. Acknowledgements We acknowledge support by the National Cancer Institute (R01CA252937 and U01CA196406), the American Cancer Society (127508-RSG-15-045-01-LIB), the Leukemia and Lymphoma Society (6551–18), the UW Trillium Myeloma Fund and the Robert J. Shillman Foundation. Ethics Approval Laboratory animal work was performed under IACUC-approved protocols #M5476 and #S19109 in the University of Wisconsin-Madison and University of California, San Diego respectively.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2022
    In:  Journal of Molecular Medicine Vol. 100, No. 9 ( 2022-09), p. 1253-1265
    In: Journal of Molecular Medicine, Springer Science and Business Media LLC, Vol. 100, No. 9 ( 2022-09), p. 1253-1265
    Type of Medium: Online Resource
    ISSN: 0946-2716 , 1432-1440
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 1462132-0
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 60, No. 10 ( 2019-08-24), p. 2558-2562
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2019
    detail.hit.zdb_id: 2030637-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 3088-3088
    Abstract: Background: Despite unprecedented response rates associated with "novel agents", HDT/AHCT is a therapeutic mainstay for transplant eligible patients with MM. However, relapse is universal and almost all patients diagnosed with symptomatic myeloma will die of their disease. The immunosuppressive network orchestrated by regulatory myeloid or lymphoid cells and effector dysfunction characterize relapses in MM. We have shown that myeloma-infiltrating myeloid cells produce versican (VCAN), a large matrix proteoglycan that promotes tumor sustaining inflammation and immunosuppression. VCAN proteolysis by a-disintegrin-and-metalloproteinase-with-thrombospondin-motifs(ADAMTS) proteases generates versikine, a bioactive fragment ("matrikine") that regulates Batf3-dendritic cells that control CD8+-attracting chemokine networks. We have shown that VCAN proteolysis predicts post AHCT survival in myeloma in a small series of patients. Here, we explore whether VCAN proteolysis is prognostic of outcomes in an expanded cohort. Methods: Bone marrow core biopsies from patients who underwent HDT/AHCT for MM from 2015-2018 were analyzed at day 90-100 (n=66). ADAMTS protease-mediated VCAN proteolysis was analyzed by immunohistochemistry from the biopsies through detection of neo-epitope DPEAAE as described previously. CD8+ T cells per high power field (HPF) was calculated at 400X magnification (10X ocular with a 40X objective) for available patients (N=35). Patients were divided into low (1+), moderate (2+) and high (3+) groups based on the scoring intensity. Patient characteristics were compared between the three groups using chi-square test or ANOVA as appropriate. Correlation analyses for VCAN proteolysis and CD8+ T cells/HPF were performed using ANOVA and linear regression in patients with available data (n=35). Kaplan Meier estimates were used to generate overall survival (OS) and progression-free survival (PFS). Multivariate analysis using Cox regression model were performed to correlate the association between VCAN proteolysis with PFS and OS using factors that affect outcomes: age, gender, cytogenetics, and treatment regimens. Logistic regression analyses were used to estimate predictors of 2-year OS and PFS. Data were analyzed using SPSS and p 〈 0.05 was considered statistically significant. Results: A total of 66 patients were included and VCAN proteolysis intensity was observed as low (1+) in 32 (48.5%), moderate (2+) in 23 (35%) and high (3+) in 11 (17%) patients respectively. Baseline characteristics between the three groups were similar (Table 1). VCAN proteolysis did not correlate with pre-AHCT response rates but was associated with post AHCT response ( 〉 very good partial response, VGPR in low: 87.5%, moderate: 65%, high: 45.5%; p=0.016, Figure 1A). The median follow-up of survivors was 37 months (range 7-116). Higher VCAN proteolysis had worse median PFS (months) [9 (95% CI 0-24) in high vs. 29 (95% CI 11-47) in moderate vs. 53 (95% CI 43-63) in low; p=0.019, Fig 1B) but no difference was observed in OS (Figure 1C). On multivariate analysis, moderate/high VCAN proteolysis had significantly worse PFS compared to low cohort (HR 2.62, 95% CI 1.20-5.74; p=0.016), but not significant for OS (HR 1.82, 95% CI 0.78-4.20; p=0.164). High/moderate VCAN proteolysis was associated with worse 2-year PFS (OR 2.79, 95% CI 1.02-7.64; p=0.046) and 2-year OS (OR 4.26, 95% CI 1.32-13.74; p=0.015) respectively. VCAN proteolysis intensity also correlated with higher CD8+ T cells (median in low: 15.5, moderate: 26, high: 66; regression coefficient 21.9, 95% CI 11.8-32.0, p 〈 0.001). Of 25 patients with VCAN status available at diagnosis, 4 (16%) converted from low to moderate/high status, while 5 (20%) converted from moderate/high to low VCAN status post AHCT respectively. Conclusions: We demonstrate in an expanded cohort that VCAN proteolysis status is associated independently with post AHCT response status and PFS. Increasing VCAN proteolysis signal intensity was associated with progressively worse outcomes consistent with the hypothesis that VCAN proteolysis is a surrogate marker for an immunosuppressive environmental network that promotes MM survival and CD8 T cell dysfunction. VCAN accumulation and turnover maybe predictive of early risk of relapse and can form the basis of future clinical trials aimed at restoring anti-MM immunity guided by VCAN proteolysis status. Disclosures Dhakal: Amgen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Honoraria; Takeda: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees. Hari:Celgene: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; BMS: Consultancy, Research Funding; Janssen: Consultancy, Honoraria; Kite: Consultancy, Honoraria; Amgen: Research Funding; Spectrum: Consultancy, Research Funding; Sanofi: Honoraria, Research Funding; Cell Vault: Equity Ownership; AbbVie: Consultancy, Honoraria.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 35, No. 15_suppl ( 2017-05-20), p. e15082-e15082
    Abstract: e15082 Background: Colorectal cancer (CRC) originates within immunologically complex microenvironments. To date the benefits of immunotherapy have been modest except in neoantigen-laden mismatch repair (MMR)-deficient tumors. Approaches to enhance tumor-infiltrating lymphocytes (TILS) in the tumor bed may substantially augment clinical immunotherapy responses. Proteolysis of the tolerogenic matrix proteoglycan versican (VCAN) generates a bioactive fragment, versikine, with putative immunostimulatory activities. Methods: Matched normal and CRC tissue samples were collected from 122 patients with cancers across all stages and locations throughout the colon and rectum. These samples were stained for VCAN, αDPEAAE (neoepitope generated in cleaving VCAN to versikine), and CD8 and scored by a pathologist. Tumors were classified as VCAN proteolysis-predominant (VPP) if their staining for total VCAN staining intensity was 〈 1+ and staining for VCAN proteolysis (αDPEAAE antibody) was 〉 2. Conversely, tumors were classified as VCAN proteolysis-weak (VPW) if intact VCAN staining intensity was 〉 1+ or αDPEAAE intensity was 〈 2+. IHC for mismatch repair (MMR) proteins was also performed. Results: Overall increased VCAN staining was observed in cancer versus (vs) normal tissue. VPP tumors had a 10 fold greater infiltration of CD8+ T-cells vs VPW cancers (p 〈 0.001). The correlation between VCAN proteolysis and CD8+ T-cell infiltration was maintained in both cancers with proficient (p) MMR and deficient (d) MMR. In both pMMR and dMMR, the VPP tumors had the greatest degree of CD8+ T-cell infiltration (Wilcoxon rank sum tests: pMMR p = 0.006; dMMR p = 0.03). Among the VPP tumors there was a greater degree of CD8+ T cell infiltration in the dMMR cancers vs pMMR cancers (35 versus 14.8 TILs per high power filed, p = 0.04). Nuclear CTNNB1, a marker for activation of WNT signaling, negatively correlated with CD8+ T cell infiltration( p = 0.014). In addition, VCAN accumulation correlated with the presence of nuclear CTNNB1 (p 〈 0.001) Conclusions: This is the first description indicating that VCAN proteolysis may shape CRC immune contexture and provide a rationale for testing VCAN proteolysis as a predictive and/or prognostic immune biomarker.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2017
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3018-3018
    Abstract: Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells (PCs) and aberrant production of monoclonal immunoglobulin detected as an M spike using serum protein electrophoresis. In the United States, MM represents ~15% of hematologic malignancies and is one of the few cancers increasing in incidence (e.g., 14,400 in 1996 to 30,330 in 2016, from SEER). Previously, a Vk*MYC mouse model was described, in which AID-dependent activation of MYC transgene in germinal center (GC) B cells catalyzes a highly penetrant, indolent MM after a prolonged latency. However, the indolent MM developed in Vk*MYC mice rarely progresses to a malignant stage, suggesting that additional genetic mutations are required for the malignant MM progression. Recent sequencing of paired tumor/normal samples from advanced or refractory MM patients identified that constitutive activation of Ras signaling pathway (KRAS: 23%; NRAS:20%; BRAF: 8%) associates with MM progression and therapy resistance. To determine whether oncogenic Nras promotes the progression of Myc-induced indolent MM to a malignant stage, we generated NrasLSL Q61R/+; Vk*MYC; IgG1-Cre (VQ) mice along with single mutant mice Vk*MYC; IgG1-Cre (Vk) or NrasLSL Q61R/+; IgG1-Cre (Q61R). To boost NrasQ61R expression in GC B cells, 6-7 weeks old mice were immunized with NP-CGG. A significant fraction of VQ mice developed M-spike after immunization and subsequently died of a highly malignant MM. The MM developed in some mice were restricted in bone marrow and spleen and thus mimicked majority of MM patients, while MM developed in other mice was primarily located in lymph nodes and represented patients with extramedullary MM. Both types of MM cells displayed hyperproliferation and hyperactivation of AKT and ERK pathways. They could be cultured for several weeks with their original stromal cells and in the presence of Il-6. The cultured MM cells could be easily infected or transfected. Upon transplantation, both types of MM cells caused a rapid lethality (within 16 and 4 weeks respectively) in secondary and tertiary recipients and preserved the initial locations of donor MM cells. MM recipient mice developed bone lesions and/or fractures characteristic of MM patients. Preliminary preclinical studies indicated that both types of MM cells were resistant to Bortezomib, a proteasome inhibitor commonly used in treating MM patients, and AZD6244, a potent MEK inhibitor. Additional tests of combinatorial therapies are ongoing. Taken together, we generated a novel mouse model in which activation of Vk*MYC and oncogenic Nras in GC B cells results in a highly malignant, transplantable MM. This model can serve as a platform to investigate pathogenesis of highly malignant multiple myeloma and test the efficacy of novel therapeutic agents. Citation Format: Zhi Wen, Adhithi Rajagopalan, Erik A. Ranheim, Remington Finn, Adam Pagenkopf, Grant Yun, Yun Zhou, Yidan Wang, Demin Wang, Marta Chesi, P. Leif Bergsagel, Fotis Asimakopoulos, Jing Zhang. Expression of oncogenic Nras and a MYC transgene in germinal center B cells induces a highly malignant multiple myeloma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 3018.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2021
    In:  The Journal of Immunology Vol. 206, No. 1_Supplement ( 2021-05-01), p. 105.01-105.01
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 206, No. 1_Supplement ( 2021-05-01), p. 105.01-105.01
    Abstract: Many autoimmune diseases feature strikingly increased prevalence in females, such as systemic lupus erythematosus (female-to-male ratio 9:1), systemic sclerosis (female-to-male ratio 11:1) and Sjögren’s syndrome (female-to-male ratio 14:1). However, the molecular basis underlying female-biased autoimmunity remains elusive. To address this knowledge gap, we performed transcriptomic profiling of minor salivary gland-derived mesenchymal stromal cells (MSCs) from primary Sjögren’s syndrome (pSS) patients and control subjects. While autosomal gene expression profiles were largely comparable between pSS and control MSCs, we detected major differences in the regulation of X-linked genes. In control female MSCs, X-linked genes that escaped inactivation were expressed from both parental and maternal X chromosomes with a median paternal ratio of ~0.5. However, in pSS female MSCs, escapees exhibited preferential expression from one of the two X chromosomes. Concomitantly, pSS MSCs showed decrease in XIST levels and reorganization of H3K27me3+ foci in the nucleus, suggesting a global dysregulation of X-inactivation maintenance. The skewing in X escape in pSS MSCs was accompanied by mislocation of protein products encoded by the escapees. Given the hallmark features of inflammation and fibrosis in pSS salivary glands and growing evidence supporting MSCs contributing to tissue homeostasis, our data suggests that dysregulation of X-inactivation maintenance, a female-specific process, contributes to the female-bias in pSS susceptibility.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2021
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 199, No. 5 ( 2017-09-01), p. 1933-1941
    Abstract: Colorectal cancer originates within immunologically complex microenvironments. To date, the benefits of immunotherapy have been modest, except in neoantigen-laden mismatch repair–deficient tumors. Approaches to enhance tumor-infiltrating lymphocytes in the tumor bed may substantially augment clinical immunotherapy responses. In this article, we report that proteolysis of the tolerogenic matrix proteoglycan versican (VCAN) strongly correlated with CD8+ T cell infiltration in colorectal cancer, regardless of mismatch repair status. Tumors displaying active VCAN proteolysis and low total VCAN were associated with robust (10-fold) CD8+ T cell infiltration. Tumor-intrinsic WNT pathway activation was associated with CD8+ T cell exclusion and VCAN accumulation. In addition to regulating VCAN levels at the tumor site, VCAN proteolysis results in the generation of bioactive fragments with novel functions (VCAN-derived matrikines). Versikine, a VCAN-derived matrikine, enhanced the generation of CD103+CD11chiMHCIIhi conventional dendritic cells (cDCs) from Flt3L-mobilized primary bone marrow–derived progenitors, suggesting that VCAN proteolysis may promote differentiation of tumor-seeding DC precursors toward IRF8- and BATF3-expressing cDCs. Intratumoral BATF3-dependent DCs are critical determinants for T cell antitumor immunity, effector T cell trafficking to the tumor site, and response to immunotherapies. Our findings provide a rationale for testing VCAN proteolysis as a predictive and/or prognostic immune biomarker and VCAN-derived matrikines as novel immunotherapy agents.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2017
    detail.hit.zdb_id: 1475085-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Oxford University Press (OUP) ; 2017
    In:  Journal of Leukocyte Biology Vol. 102, No. 2 ( 2017-03-02), p. 265-275
    In: Journal of Leukocyte Biology, Oxford University Press (OUP), Vol. 102, No. 2 ( 2017-03-02), p. 265-275
    Abstract: The last 10–15 years have witnessed a revolution in treating multiple myeloma, an incurable cancer of Ab-producing plasma cells. Advances in myeloma therapy were ushered in by novel agents that remodel the myeloma immune microenvironment. The first generation of novel agents included immunomodulatory drugs (thalidomide analogs) and proteasome inhibitors that target crucial pathways that regulate immunity and inflammation, such as NF-κB. This paradigm continued with the recent regulatory approval of mAbs (elotuzumab, daratumumab) that impact both tumor cells and associated immune cells. Moreover, recent clinical data support checkpoint inhibition immunotherapy in myeloma. With the success of these agents has come the growing realization that the myeloid infiltrate in myeloma lesions—what we collectively call the myeloid-in-myeloma compartment—variably sustains or deters tumor cells by shaping the inflammatory milieu of the myeloma niche and by promoting or antagonizing immune-modulating therapies. The myeloid-in-myeloma compartment includes myeloma-associated macrophages and granulocytes, dendritic cells, and myeloid-derived-suppressor cells. These cell types reflect variable states of differentiation and activation of tumor-infiltrating cells derived from resident myeloid progenitors in the bone marrow—the canonical myeloma niche—or myeloid cells that seed both canonical and extramedullary, noncanonical niches. Myeloma-infiltrating myeloid cells engage in crosstalk with extracellular matrix components, stromal cells, and tumor cells. This complex regulation determines the composition, activation state, and maturation of the myeloid-in-myeloma compartment as well as the balance between immunogenic and tolerogenic inflammation in the niche. Redressing this balance may be a crucial determinant for the success of antimyeloma immunotherapies.
    Type of Medium: Online Resource
    ISSN: 0741-5400 , 1938-3673
    RVK:
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2017
    detail.hit.zdb_id: 2026833-6
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages