scholarly journals Regulation of CNS Lymphoma Progression By the Myeloid Microenvironment

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 448-448
Author(s):  
Hua-Xin Gao ◽  
Eleanor Fraser ◽  
Mario Merlini ◽  
Huimin Geng ◽  
Lingjing Chen ◽  
...  

Abstract ;'Insights into the molecular and immunologic pathogenesis of primary CNS lymphomas are essential for meaningful progress in therapy. Tumor-associated macrophages represent the dominant infiltrating leukocyte and there are few established insights into their phenotypes and role in this disease. While upregulation of Th2 cytokines IL-4 and IL-10 in the microenvironment has been demonstrated, the relative roles of M1 and M2 macrophages in contributing to CNS lymphoma pathogenesis has not been elucidated. To date, there is also no information regarding the relative contributions of brain resident microglia and infiltrating macrophages and their interactions with lymphoma. Additional key questions include the identification of factors that mediate both immune cell chemotaxis in CNS lymphomas, as well as the relationship between myeloid cell infiltration and T-cell mediated immune surveillance and immunosuppression. We combined analyses of clinical specimens and mechanistic studies using preclinical in vivo models and show evidence that infiltrating tumor-associated macrophages, derived from monocyte precursors, have a critical role in attenuating CNS lymphoma progression. Immunohistochemical analysis of the density and morphologic features of CD68+ tumor-associated macrophages in 62 diagnostic specimens of immunocompetent PCNSL demonstrated that smaller macrophage size and lower macrophage density correlated with significantly shorter OS. Evaluation of CD68 immunoreactivity using image analysis software (ImageJ) confirmed the heterogeneity of macrophage size and infiltrative density in PCNSL. A multivariate Cox model including age, IELSG score, receipt of consolidation and/or maintenance therapy demonstrated that tumor-associated macrophage density (both count and area) was a significant, independent predictor of favorable PFS and OS and that larger macrophage size a significant, independent predictor of OS in PCNSL treated with standard MTX-based induction (predominantly MTX, temozolomide, rituximab). Using a variety of syngeneic and non-syngeneic preclinical models, including patient-derived CNS lymphoma cells, as well as diagnostic clinical specimens, we characterized the phenotype of tumor-associated macrophages in PCNSL. Using flow-cytometry, we demonstrated that while CD45 high tumor-associated macrophages exhibit strong expression of the canonical M2 marker CD206, a scavenger receptor, these also displayed high co-expression of iNOS and MHC II, markers of classically-activated M1 macrophages. Pharmacologic inhibition of the CSF-1 receptor led to accelerated CNS lymphoma progression, attenuated T-cell infiltration and blocked rituximab efficacy. A flow-cytometric assay of phagocytosis, using Raji lymphoma transduced to express mCherry, demonstrated that infiltrating CD206+ macrophages are the dominant mediator of lymphoma phagocytosis. We applied 2P intravital imaging of a CNS lymphoma model using Cx3cr1GFP/+:Ccr2RFP/+ myeloid cell dual reporter mice and transcriptional studies to define the time-dependent infiltration and phenotypic changes in tumor-associated macrophages and microglia that correlate with disease progression. Using IFN-γ -/- mice we identified a critical role for IFN-γ in the regulation of CNS lymphoma, in the presence and absence of T-cells. We identified IFN-γ-regulated genes in tumor-associated macrophages that may contribute to direct lymphoma cytotoxicity as well as stimulation of T-cell chemotaxis and antigen processing, including TAP1 and TAP2. By IHC, we confirmed TAP1 expression in a subset of diagnostic specimens of PCNSL and determined, using Cox multivariate model, that strong TAP1 correlated with improved PFS (p<0.0006). Notably, independent of receipt of maintenance therapy, TAP1 also correlated with improved PFS in 38 patients that received only MTX-based induction, without dose-intensive chemotherapy consolidation. (Figure 1) Our results support a direct, immune-editing role for monocyte-derived macrophages in the regulation of CNS lymphoma progression, via several mechanisms, including antigenic processing and cross-presentation. We suggest that tumor-associated CD68 and TAP1 (and TAP2) be evaluated further as candidate biomarkers for risk stratification in PCNSL, particularly in trials that involve targeted immunotherapy. Supported by NCI and LLS. Figure 1 Figure 1. Disclosures Rubenstein: Kymera: Research Funding.

Blood ◽  
2003 ◽  
Vol 102 (8) ◽  
pp. 2933-2939 ◽  
Author(s):  
Paola Bernabei ◽  
Marita Bosticardo ◽  
Giuliana Losana ◽  
Gabriella Regis ◽  
Francesca Di Paola ◽  
...  

Abstract The ability of insulin-like growth factor-1 (IGF-1) to regulate surface expression of the interferon-γ receptor 2 (IFN-γR2) transducing chain and activation of IFN-γ–induced signal transducer and activator of transcription-1 (STAT-1) in human T cells was analyzed. We show that, especially in the absence of serum (which contains IGF-1), IGF-1 down-regulated surface expression of the IFN-γR2 chain and inhibited both IFN-γ–dependent STAT-1 activation and apoptosis in T-cell lines ST4, Jurkat, and Molt-4. IFN-γR2 down-regulation resulted from its enhanced internalization since IGF-1 completely restored the uptake of anti–IFN-γR2 monoclonal antibody (mAb) in serum-deprived T-cell lines. When the interaction between IGF-1 and its receptor was blocked by anti–IGF-1R mAb, enhancement of IFN-γR2 surface expression, STAT-1 activation, and reinstatement of IFN-γ–induced apoptosis were observed. Enhanced expression of IFN-γR2 was also observed in phytohemagglutinin (PHA)–activated T lymphoblasts cultured in the presence of anti–IGF-1R mAb, whereas IGF-1 or anti–IGF-1R mAb did not modify the high IFN-γR2 expression in B and myeloid cell lines. Both IGF-1 and anti–IGF-1R mAb did not modify the constitutive expression of IFN-γR2 mRNA in T cells as well as the high IFN-γR1 binding chain surface expression in T, B, and myeloid cells. These data indicate that IGF-1 plays a critical role in the desensitization of IFN-γ/STAT-1 signaling in T lymphocytes by delivering a signal for IFN-γR2 internalization.


2021 ◽  
Vol 6 (56) ◽  
pp. eabb9435
Author(s):  
Joseph M. Leal ◽  
Jessica Y. Huang ◽  
Karan Kohli ◽  
Caleb Stoltzfus ◽  
Miranda R. Lyons-Cohen ◽  
...  

Microanatomical organization of innate immune cells within lymph nodes (LNs) is critical for the generation of adaptive responses. In particular, steady-state LN-resident dendritic cells (Res cDCs) are strategically localized to intercept lymph-draining antigens. Whether myeloid cell organization changes during inflammation and how that might affect the generation of immune responses are unknown. Here, we report that during type I, but not type II, inflammation after adjuvant immunization or viral infection, antigen-presenting Res cDCs undergo CCR7-dependent intranodal repositioning from the LN periphery into the T cell zone (TZ) to elicit T cell priming. Concurrently, inflammatory monocytes infiltrate the LNs via local blood vessels, enter the TZ, and cooperate with Res cDCs by providing polarizing cytokines to optimize T cell effector differentiation. Monocyte infiltration is nonuniform across LNs, generating distinct microenvironments with varied local innate cell composition. These spatial microdomains are associated with divergent early T cell effector programming, indicating that innate microenvironments within LNs play a critical role in regulating the quality and heterogeneity of T cell responses. Together, our findings reveal that dynamic modulation of innate cell microenvironments during type I inflammation leads to optimized generation of adaptive immune responses to vaccines and infections.


2005 ◽  
Vol 202 (2) ◽  
pp. 231-237 ◽  
Author(s):  
Indira Guleria ◽  
Arezou Khosroshahi ◽  
Mohammed Javeed Ansari ◽  
Antje Habicht ◽  
Miyuki Azuma ◽  
...  

Fetal survival during gestation implies that tolerance mechanisms suppress the maternal immune response to paternally inherited alloantigens. Here we show that the inhibitory T cell costimulatory molecule, programmed death ligand 1 (PDL1), has an important role in conferring fetomaternal tolerance in an allogeneic pregnancy model. Blockade of PDL1 signaling during murine pregnancy resulted in increased rejection rates of allogeneic concepti but not syngeneic concepti. Fetal rejection was T cell– but not B cell–dependent because PDL1-specific antibody treatment caused fetal rejection in B cell–deficient but not in RAG-1–deficient females. Blockade of PDL1 also resulted in a significant increase in the frequency of IFN-γ–producing lymphocytes in response to alloantigen in an ELISPOT assay and higher IFN-γ levels in placental homogenates by ELISA. Finally, PDL1-deficient females exhibited decreased allogeneic fetal survival rates as compared with littermate and heterozygote controls and showed evidence of expansion of T helper type 1 immune responses in vivo. These results provide the first evidence that PDL1 is involved in fetomaternal tolerance.


Blood ◽  
2012 ◽  
Vol 120 (16) ◽  
pp. 3326-3335 ◽  
Author(s):  
Hui Zhong ◽  
Weili Bao ◽  
Xiaojuan Li ◽  
Allison Miller ◽  
Caroline Seery ◽  
...  

Abstract Immune thrombocytopenia (ITP) results from decreased platelet production and accelerated platelet destruction. Impaired CD4+ regulatory T-cell (Treg) compartment and skewed Th1 and possibly Th17 responses have been described in ITP patients. The trigger for aberrant T-cell polarization remains unknown. Because monocytes have a critical role in development and polarization of T-cell subsets, we explored the contribution of monocyte subsets in control of Treg and Th development in patients with ITP. Unlike circulating classic CD14hiCD16− subpopulation, the CD16+ monocyte subset was expanded in ITP patients with low platelet counts on thrombopoietic agents and positively correlated with T-cell CD4+IFN-γ+ levels, but negatively with circulating CD4+CD25hiFoxp3+ and IL-17+ Th cells. Using a coculture model, we found that CD16+ ITP monocytes promoted the expansion of IFN-γ+CD4+ cells and concomitantly inhibited the proliferation of Tregs and IL-17+ Th cells. Th-1–polarizing cytokine IL-12, secreted after direct contact of patient T-cell and CD16+ monocytes, was responsible for the inhibitory effect on Treg and IL-17+CD4+ cell proliferation. Our findings are consistent with ITP CD16+ monocytes promoting Th1 development, which in turn negatively regulates IL-17 and Treg induction. This underscores the critical role of CD16+ monocytes in the generation of potentially pathogenic Th responses in ITP.


2002 ◽  
Vol 76 (9) ◽  
pp. 4497-4506 ◽  
Author(s):  
Catherine E. Patterson ◽  
Diane M. P. Lawrence ◽  
Lisa A. Echols ◽  
Glenn F. Rall

ABSTRACT Neurons of the mammalian central nervous system (CNS) are an essential and largely nonrenewable cell population. Thus, virus infections that result in neuronal depletion, either by virus-mediated cell death or by induction of the cytolytic immune response, could cause permanent neurological impairment of the host. In a transgenic mouse model of measles virus (MV) infection of neurons, we have previously shown that the host T-cell response was required for resolution of infection in susceptible adult mice. In this report, we show that this protective response did not result in neuronal death, even during the peak of T-cell infiltration into the brain parenchyma. When susceptible mice were intercrossed with specific immune knockout mice, a critical role for gamma interferon (IFN-γ) was identified in protection against MV infection and CNS disease. Moreover, the addition of previously activated splenocytes or recombinant murine IFN-γ to MV-infected primary neurons resulted in the inhibition of viral replication in the absence of neuronal death. Together, these data support the hypothesis that the host immune response can promote viral clearance without concomitant neuronal loss, a process that appears to be mediated by cytokines.


2008 ◽  
Vol 76 (8) ◽  
pp. 3628-3631 ◽  
Author(s):  
Sumana Chakravarty ◽  
G. Christian Baldeviano ◽  
Michael G. Overstreet ◽  
Fidel Zavala

ABSTRACT The protective immune response against liver stages of the malaria parasite critically requires CD8+ T cells. Although the nature of the effector mechanism utilized by these cells to repress parasite development remains unclear, a critical role for gamma interferon (IFN-γ) has been widely assumed based on circumstantial evidence. However, the requirement for CD8+ T-cell-mediated IFN-γ production in protective immunity to this pathogen has not been directly tested. In this report, we use an adoptive transfer strategy with circumsporozoite (CS) protein-specific transgenic T cells to examine the role of CD8+ T-cell-derived IFN-γ production in Plasmodium yoelii-infected mice. We show that despite a marginal reduction in the expansion of naive IFN-γ-deficient CS-specific transgenic T cells, their antiparasite activity remains intact. Further, adoptively transferred IFN-γ-deficient CD8+ T cells were as efficient as their wild-type counterparts in limiting parasite growth in naive mice. Taken together, these studies demonstrate that IFN-γ secretion by CS-specific CD8+ T cells is not essential to protect mice against live sporozoite challenge.


2014 ◽  
Vol 82 (10) ◽  
pp. 4092-4103 ◽  
Author(s):  
Abinav Kumar Singh ◽  
Nagaraja R. Thirumalapura

ABSTRACTDiverse pathogens have evolved to survive and replicate in the endosomes or phagosomes of the host cells and establish persistent infection. Ehrlichiae are Gram-negative, intracellular bacteria that are transmitted by ticks. Ehrlichiae reside in the endosomes of the host phagocytic or endothelial cells and establish persistent infection in their vertebrate reservoir hosts. CD4+T cells play a critical role in protection against phagosomal infections. In the present study, we investigated the expansion, maintenance, and functional status of antigen-specific CD4+T cells during persistentEhrlichia murisinfection in wild-type and interleukin-10 (IL-10)-deficient mice. Our study indicated that early induction of IL-10 led to reduced inflammatory responses and impaired bacterial clearance during persistentEhrlichiainfection. Notably, we demonstrated that the functional production of gamma interferon (IFN-γ) by antigen-specific CD4+T cells maintained during a persistent phagosomal infection progressively deteriorates. The functional loss of IFN-γ production by antigen-specific CD4+T cells was reversed in the absence of IL-10. Furthermore, we demonstrated that transient blockade of IL-10 receptor during the T cell priming phase early in infection was sufficient to enhance the magnitude and the functional capacity of antigen-specific effector and memory CD4+T cells, which translated into an enhanced recall response. Our findings provide new insights into the functional status of antigen-specific CD4+T cells maintained during persistent phagosomal infection. The study supports the concept that a better understanding of the factors that influence the priming and differentiation of CD4+T cells may provide a basis to induce a protective immune response against persistent infections.


Blood ◽  
2012 ◽  
Vol 119 (1) ◽  
pp. 127-136 ◽  
Author(s):  
Min Chen ◽  
Kumar Felix ◽  
Jin Wang

AbstractAfter stimulation of antigen-specific T cells, dendritic cell (DCs) are susceptible to killing by these activated T cells that involve perforin and Fas-dependent mechanisms. Fas-dependent DC apoptosis has been shown to limit DC accumulation and prevent the development of autoimmunity. However, a role for perforin in the maintenance of DC homeostasis for immune regulation remains to be determined. Here we show that perforin deficiency in mice, together with the deletion of Fas in DCs (perforin−/−DC-Fas−/−), led to DC accumulation, uncontrolled T-cell activation, and IFN-γ production by CD8+ T cells, resulting in the development of lethal hemophagocytic lymphohistiocytosis. Consistently, adoptive transfer of Fas−/− DCs induced over-activation and IFN-γ production in perforin−/− CD8+ T cells. Neutralization of IFN-γ prevented the spreading of inflammatory responses to different cell types and protected the survival of perforin−/−DC-Fas−/− mice. Our data suggest that perforin and Fas synergize in the maintenance of DC homeostasis to limit T cell activation, and prevent the initiation of an inflammatory cascade.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 374-374 ◽  
Author(s):  
Hua-Xin Gao ◽  
Shawn Anderson ◽  
Cigall Kadoch ◽  
Mekhala Maiti ◽  
Lingjing Chen ◽  
...  

Abstract Background CNS manifestations of aggressive non-Hodgkin Lymphoma are associated with serious morbidity and adverse prognosis. Primary CNS lymphomas (PCNSL) exhibit a dichomatous growth pattern, either dissemination within brain, typical at presentation, and/or leptomeningeal spread, common at relapse. Elucidation of the mechanistic basis of CNS lymphoma progression as well as drug resistance requires preclinical models that recapitulate their pathogenesis. Methods We developed a novel method to derive cell lines of CNS lymphoma that recapitulate disease phenotypes upon intracranial implantation into mice. We are applying genomics, in vitrochemotaxis, preclinical testing of targeted therapies and neuroimaging to evaluate mechanisms of invasion and resistance. Results We developed 7 CNS lymphoma cell lines; 6 DLBCL (all ABC-type), 1 Burkitt; 5 from secondary CNS lymphoma (SCNSL), and 2 from PCNSL, of which 1 was treatment naïve. Intracranial implantation of lymphoma cells from these tumours within NSG mice provides a reproducible model to dissect the pathogenesis of CNS lymphomas. PCNSL specimens were 10X more efficient in CNS dissemination than SCNSL. High resolution array-CGH demonstrated that intracranial tumour growth was associated with retention of genomic aberrations of the original tumours (e.g. del 6q, gains on 12, etc) and that these were maintained with serial passage in vivo. CNS-infiltrative lymphomas expressed significantly increased levels of MMP-7 and RGS-13 transcripts compared to lymphomas that did not infiltrate brain, while osteopontin and cathepsin D expression by lymphoma cells did not correlate with CNS invasion. Targeted shRNA-mediated knockdown of RGS-13 was performed using lentiviral infection and resulted in significant delay of CNS lymphoma growth in vivo in a xenograft model but had no effect on lymphoma proliferation in culture. Therapeutic response to lenalidomide, minus and plus rituximab, was recapitulated in RAG-/- mice, despite deficient T-cell function and correlated with baseline relative cereblon expression, as quantified using a highly specific immunohistochemical assay. The emergence of resistance to lenalidomide in human CNS lymphoma xenografts also correlated with loss of cereblon protein expression, supporting a role for cereblon in the efficacy of lenalidomide in CNS lymphomas. Notably, significant cereblon protein expression by lymphoma cells was detected by immunohistochemistry in 12/22 diagnostic specimens of aggressive CNS lymphoma. Metabolic imaging of model CNS lymphomas using magnetic resonance spectroscopy demonstrated significant intratumoural lactate production in the microenvironment, detectable before evidence of aberrant T2 signal and reduced diffusion. Lenalidomide reduced tumour expression of lactate dehydrogenase and lactate, as well as RGS-13, consistent with anti-proliferative as well as anti-invasive effects. Conclusions To the best of our knowledge we have developed the first panel of patient-derived CNS lymphoma cell lines. We have used these to generate intracranial xenografts that provide a highly reproducible model system to dissect key elements of CNS lymphoma pathogenesis, leading to the elucidation of mechanisms of CNS lymphoma growth and invasion as well as resistance. Our results support a direct, T-cell independent effect of lenalidomide on CNS lymphoma growth and invasion which may be cereblon-dependent. Additional studies are needed to define the role of cereblon as a biomarker and mediator of lenalidomide efficacy in CNS lymphomas. In addition, we are using these models to identify novel genomic and metabolic aberrations predictive of early resistance to lenalidomide and other targeted therapies. Supported by the Lymphoma Research Foundation, Leukemia and Lymphoma Society, and by NIH R01CA139-83-01A1. Disclosures: Heise: Celgene: Employment, Equity Ownership. Rubenstein:Celgene: Research Funding; Genentech: Research Funding. Off Label Use: use of lenalidomide in CNS lymphoma.


Sign in / Sign up

Export Citation Format

Share Document