Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Joseph Nthale is active.

Publication


Featured researches published by Joseph Nthale.


Science | 2016

Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity

Rodney Dixon Dorand; Joseph Nthale; Jay Myers; Deborah Barkauskas; Stefanie Avril; Steven M. Chirieleison; Tej K. Pareek; Duncan Stearns; John J. Letterio; Alex Y. Huang; Agne Petrosiute

Cyclin suppresses antitumor immunity Despite the dramatic success of cancer immunotherapy, many types of cancer do not respond. Understanding why could help us to find ways to enhance the overall responsiveness of tumors to immunotherapies. Dorand et al. report that cyclin-dependent kinase 5 (Cdk5), an enzyme that is highly expressed by neurons in many brain cancers, may dampen the ability of T cells to reject tumors. In a mouse model of medulloblastoma, if tumors were Cdk5 deficient, T cells were able to remove them. This heightened antitumor immunity correlated with reduced expression of the inhibitory molecule programmed cell death ligand 1 (PD-L1), a target of current cancer immunotherapies. Science, this issue p. 399 Cyclin-dependent kinase 5 expression by tumors inhibits antitumor immunity in human and mouse medulloblastoma. Cancers often evade immune surveillance by adopting peripheral tissue– tolerance mechanisms, such as the expression of programmed cell death ligand 1 (PD-L1), the inhibition of which results in potent antitumor immunity. Here, we show that cyclin-dependent kinase 5 (Cdk5), a serine-threonine kinase that is highly active in postmitotic neurons and in many cancers, allows medulloblastoma (MB) to evade immune elimination. Interferon-γ (IFN-γ)–induced PD-L1 up-regulation on MB requires Cdk5, and disruption of Cdk5 expression in a mouse model of MB results in potent CD4+ T cell–mediated tumor rejection. Loss of Cdk5 results in persistent expression of the PD-L1 transcriptional repressors, the interferon regulatory factors IRF2 and IRF2BP2, which likely leads to reduced PD-L1 expression on tumors. Our finding highlights a central role for Cdk5 in immune checkpoint regulation by tumor cells.


Cancer Research | 2016

Aberrant Notch Signaling in the Bone Marrow Microenvironment of Acute Lymphoid Leukemia Suppresses Osteoblast-Mediated Support of Hematopoietic Niche Function

Weihuan Wang; Grant Zimmerman; Xiaoran Huang; Shuiliang Yu; Jay Myers; Yiwei Wang; Stephen Moreton; Joseph Nthale; Amad Awadallah; Rose C. Beck; Wei Xin; David Wald; Alex Y. Huang; Lan Zhou

More than half of T-cell acute lymphoblastic leukemia (T-ALL) patients harbor gain-of-function mutations in the intracellular domain of Notch1. Diffuse infiltration of the bone marrow commonly occurs in T-ALL and relapsed B-cell acute lymphoblastic leukemia patients, and is associated with worse prognosis. However, the mechanism of leukemia outgrowth in the marrow and the resulting biologic impact on hematopoiesis are poorly understood. Here, we investigated targetable cellular and molecular abnormalities in leukemia marrow stroma responsible for the suppression of normal hematopoiesis using a T-ALL mouse model and human T-ALL xenografts. We found that actively proliferating leukemia cells inhibited normal hematopoietic stem and progenitor cell (HSPC) proliferation and homing to the perivascular region. In addition, leukemia development was accompanied by the suppression of the endosteum-lining osteoblast population. We further demonstrated that aberrant Notch activation in the stroma plays an important role in negatively regulating the expression of CXLC12 on osteoblasts and their differentiation. Notch blockade reversed attenuated HSPC cycling, leukemia-associated abnormal blood lineage distribution, and thrombocytopenia as well as recovered osteoblast and HSPC abundance and improved the hematopoietic-supportive functions of osteoblasts. Finally, we confirmed that reduced osteoblast frequency and enhanced Notch signaling were also features of the marrow stroma of human ALL tissues. Collectively, our findings suggest that therapeutically targeting the leukemia-infiltrated hematopoietic niche may restore HSPC homeostasis and improve the outcome of ALL patients.


Journal of Immunology | 2016

Transient Surface CCR5 Expression by Naive CD8+ T Cells within Inflamed Lymph Nodes Is Dependent on High Endothelial Venule Interaction and Augments Th Cell–Dependent Memory Response

David Askew; Charles A. Su; Deborah Barkauskas; R. Dixon Dorand; Jay Myers; Rachel Liou; Joseph Nthale; Alex Y. Huang

In inflamed lymph nodes, Ag-specific CD4+ and CD8+ T cells encounter Ag-bearing dendritic cells and, together, this complex enhances the release of CCL3 and CCL4, which facilitate additional interaction with naive CD8+ T cells. Although blocking CCL3 and CCL4 has no effect on primary CD8+ T cell responses, it dramatically impairs the development of memory CD8+ T cells upon Ag rechallenge. Despite the absence of detectable surface CCR5 expression on circulating native CD8+ T cells, these data imply that naive CD8+ T cells are capable of expressing surface CCR5 prior to cognate Ag-induced TCR signaling in inflamed lymph nodes; however, the molecular mechanisms have not been characterized to date. In this study, we show that CCR5, the receptor for CCL3 and CCL4, can be transiently upregulated on a subset of naive CD8+ T cells and that this upregulation is dependent on direct contact with the high endothelial venule in inflamed lymph node. Binding of CD62L and CD11a on T cells to their ligands CD34 and CD54 on the high endothelial venule can be enhanced during inflammation. This enhanced binding and subsequent signaling promote the translocation of CCR5 molecules from intracellular vesicles to the surface of the CD8+ T cell. The upregulation of CCR5 on the surface of the CD8+ T cells increases the number of contacts with Ag-bearing dendritic cells, which ultimately results in increased CD8+ T cell response to Ag rechallenge.


Frontiers in Immunology | 2017

CCL3 Enhances Antitumor Immune Priming in the Lymph Node via IFNγ with Dependency on Natural Killer Cells

Frederick Allen; Peter Rauhe; David Askew; Alexander Tong; Joseph Nthale; Saada Eid; Jay Myers; Caryn Tong; Alex Y. Huang

Lymph node (LN) plays a critical role in tumor cell survival outside of the primary tumor sites and dictates overall clinical response in many tumor types (1, 2). Previously, we and others have demonstrated that CCL3 plays an essential role in orchestrating T cell—antigen-presenting cell (APC) encounters in the draining LN following vaccination, and such interactions enhance the magnitude of the memory T cell pool (3–5). In the current study, we investigate the cellular responses in the tumor-draining lymph nodes (TDLNs) of a CCL3-secreting CT26 colon tumor (L3TU) as compared to wild-type tumor (WTTU) during the priming phase of an antitumor response (≤10 days). In comparison to WTTU, inoculation of L3TU resulted in suppressed tumor growth, a phenomenon that is accompanied by altered in vivo inflammatory responses on several fronts. Autologous tumor-derived CCL3 (aCCL3) secretion by L3TU bolstered the recruitment of T- and B-lymphocytes, tissue-migratory CD103+ dendritic cells (DCs), and CD49b+ natural killer (NK) cells, resulting in significant increases in the differentiation and activation of multiple Interferon-gamma (IFNγ)-producing leukocytes in the TDLN. During this early phase of immune priming, NK cells constitute the major producers of IFNγ in the TDLN. CCL3 also enhances CD8+ T cell proliferation and differentiation by augmenting DC capacity to drive T cell activation in the TDLN. Our results revealed that CCL3-dependent IFNγ production and CCL3-induced DC maturation drive the priming of effective antitumor immunity in the TDLN.


OncoImmunology | 2018

Systemic administration of β-glucan of 200 kDa modulates melanoma microenvironment and suppresses metastatic cancer

Mei Zhang; Liane Chun; Victor Sandoval; Hallie Graor; Jay Myers; Joseph Nthale; Peter Rauhe; Zachary Senders; Kevin Choong; Alex Y. Huang; Julian Kim

ABSTRACT Converting an immunosuppressive melanoma microenvironment into one that favors the induction of antitumor immunity is indispensable for effective cancer immunotherapy. In the current study we demonstrate that oat-derived β-(1-3)-(1-4)-glucan of 200 kDa molecular size (BG34-200) previously shown to mediate direct interaction with macrophages could alter the immune signature within melanoma microenvironment. Systemic administration of BG34-200 resulted in reversion of tolerant melanoma microenvironment to an immunogenic one that allows M1-type activation of macrophages, the induction of pro-inflammatory cytokines/chemokines including IFN-γ, TNF-α, CXCL9, and CXCL10, and enhanced IRF1 and PD-L1 expression. In turn, BG34-200 induced a superior antitumor response against primary and lung metastatic B16F10 melanoma compared to untreated controls. The enhanced tumor destruction was accompanied with significantly increased tumor infiltration of CD4+ and CD8+ T cells as well as elevated IFN-γ in the tumor sites. Systemic administration of BG34-200 also provoked systemic activation of tumor draining lymph node T cells that recognize antigens naturally expressing in melanoma (gp100/PMEL). Mechanistic studies using CD11b-knockout (KO), CD11 c-DTR transgenic mice and nude mice revealed that macrophages, DCs, T cells and NK cells were all required for the BG34-200-induced therapeutic benefit. Studies using IFN-γ-KO transgenic mice showed that IFN-γ was essential for the BG34-200-elicited antitumor response. Beyond melanoma, the therapeutic efficacy of BG34-200 and its immune stimulating activity were demonstrated in a mouse model of osteosarcoma. Together, BG34-200 is highly effective in modulating antitumor immunity. Our data support the potential therapeutic use of this novel immune modulator in the treatment of metastatic melanoma.


Cancer immunology research | 2016

Abstract A050: Cdk5 disruption attenuates tumor PD-L1 expression via regulation of IFN-γ signaling components and promotes antitumor immunity

Rodney Dixon Dorand; Joseph Nthale; Jay Myers; Deborah Barkauskas; Stefanie Avril; Steven M. Chirieleison; Tej K. Pareek; Duncan Stearns; John J. Letterio; Alex Y. Huang; Agne Petrosiute

In order to escape normal immune surveillance, tumors mimic peripheral tissue tolerance mechanisms such as the expression of programmed cell death-ligand 1 (PD-L1), the inhibition of which can lead to potent anti-tumor responses. Here we show the contribution of cyclin dependent kinase 5 (Cdk5) to immune evasion. Cdk5 is a proline-directed serine/threonine kinase, which is highly expressed in post-mitotic neurons and directs a variety of homeostatic functions from cytoskeletal rearrangement to neurotransmitter signaling. Studies in xenograft models have linked Cdk5 activity to the generation and metabolic activity of primary tumors within the central nervous system (CNS). Here we show that Cdk5, expressed in both murine and human medulloblastoma (MB) cell lines, plays a major role in the ability of MB to avoid immune detection. First, we observed that decreased Cdk5 expression was associated with an increase in the number of infiltrating CD3+ cells in the tumor mass of clinical MB samples. Additionally, using publicly available datasets, we found that decreased Cdk5 expression is correlated with better overall survival or fewer distant metastasis in melanoma, breast cancer, glioma, and lung adenocarcinoma. Next, using a CRISPR/Cas-9 approach, we silenced Cdk5 in a murine model of Sonic Hedgehog (SHH) pathway MB. Rather than causing intrinsic growth defects in MB, interference of Cdk5 activity sensitizes tumors to killing by the normal host immune system in a CD4+ T cell-dependent manner. This rejection is associated with increased IFN-γ expression in the tumor microenvironment, as well as increased PD-L1 expression by myeloid populations in both subcutaneous and intracranial tumors. Mechanistically, we observed an attenuated response to IFN-γ stimulated expression of programmed death ligand 1 (PD-L1) on MB cells. This blunted response was recapitulated when MB cells were treated with Roscovitine, a non-selective Cdk5 inhibitor. Furthermore, using a quantitative global phosphoproteomics approach, we found that Cdk5 knockdown also increased phosphorylation of S-440 and S-443 on interferon regulatory factor binding protein 2 (IRF2BP2), an upstream co-repressor of interferon regulatory factor 2 (IRF-2). Increased phosphorylation of IRF2BP2 corresponded with stable expression of IRF-2 with a concomitant decrease in surface expression of PD-L1. These observations highlight a critical role for Cdk5 in the immune escape mechanisms of primary CNS tumors and provides new therapeutic targets for PD-1/PD-L1 directed immunotherapy. Citation Format: Rodney D. Dorand, Jr., Joseph Nthale, Jay T. Myers, Deborah S. Barkauskas, Stefanie Avril, Steven M. Chirieleison, Tej K. Pareek, Derek W. Abbott, Duncan S. Stearns, John J. Letterio, Alex Y. Huang, Agne Petrosiute. Cdk5 disruption attenuates tumor PD-L1 expression via regulation of IFN-γ signaling components and promotes antitumor immunity [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A050.


Cancer immunology research | 2016

Abstract B158: Cyclin dependent kinase-5 regulates IFNγ induced PD-L1 expression via IRF-1 in medulloblastoma

Rodney Dixon Dorand; Agne Petrosiute; Joseph Nthale; Tej K. Pareek; Francesca Scrimieri; Jay Myers; Deborah Barkauskas; John J. Letterio; Duncan Stearns; Alex Y. Huang

Immune checkpoint blockade offers an exciting, new adjuvant therapy to both solid and liquid tumors. However, the underlying mechanisms by which tumors alter their expression of immune checkpoint blockade ligands, such as programmed death ligand 1 (PD-L1), to achieve adaptive immune resistance needs to be further studied. We sought to investigate the mechanisms by which PD-L1 expression is regulated in response to IFNγ stimulation in medulloblastoma, a primary CNS neoplasm. MB is the most common pediatric CNS malignancy that is capable of metastasis to distant sites. MB is classified into four different molecular subgroups. Patched (PTCH), a signaling molecule of the Sonic Hedgehog (SHH) pathway, represents one subgroup and accounts for 25% of all sporadic human MB. Because MB develops from primitive cerebellar granule neuron precursor cells, it is thought that errors in development or migration of these neurons contribute to tumorigenesis. Cyclin Dependent Kinase 5 (CDK5) is a non-canonical member of the CDK family that does not require cyclin for activity but relies on binding to its obligate activators, p35 and p39 which are mostly expressed in post mitotic neurons. CDK5 is required for normal Central Nervous System (CNS) development. Utilizing both short hairpin RNA (shRNA) approach and the bacterial CRISPR-Cas9 (Cr) system we silenced Cdk5 expression in a C57BL/6 (B6) syngeneic MB line derived from p53-/- PTCH+/- (MM1). In vitro proliferation experiments revealed that MM1-WT, MM1 cells transduced with shRNA targeting CDK5 (MM1-shCDK5), and MM1 cells transduced with non-silencing, scrambled RNA sequences (MM1-shNS) had similar growth kinetics. Since there were no intrinsic differences in in vitro growth kinetics, we subcutaneously injected immunocompetent C57BL/B6 mice with 5x104 tumor cells and measured tumor growth every three days. In our first experiment, 19 days after injection 7.7% (N=13) of mice injected with MM1-WT and 0% (N=7) injected with MM1-shNS were tumor free compared with 76.5% (N=13) of animals injected with MM1-shCDK5 Comparison of tumor free survival revealed statistically significant differences between the MM1-WT with MM1-shCDK5 groups (p In summary, we have shown that Cdk5 is expressed in Type 2 MB and that silencing of its expression with shRNA results in an altered growth phenotype in vivo but not in vitro. Furthermore, eliminating the ability of MM1 tumors to express PD-L1 might lead to effective immune rejection of MM1-shCDK5. Mechanistically, we showed that this enhanced tumor rejection may be due in part to the failure of Cdk5 deficient MB cells to alter IRF-1 expression which serves as a transcription factor for PD-L1. These novel findings point to CDK5 as a potential immunotherapeutic target in MB. Citation Format: Rodney Dixon Dorand, Jr., Agne Petrosiute, Joseph Nthale, Tej Pareek, Francesca Scrimieri, Jay Myers, Deborah Barkauskas, John J. Letterio, Duncan S. Stearns, Alex Y. Huang. Cyclin dependent kinase-5 regulates IFNγ induced PD-L1 expression via IRF-1 in medulloblastoma. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B158.


Cancer immunology research | 2016

Abstract A119: CCL3 in the tumor microenvironment augments antitumor immune priming in the lymph node

Frederick Allen; Joseph Nthale; Saada Eid; Peter Rauhe; David Askew; Jay Myers; Alexander Tong; Alex Y. Huang

Any therapeutic approach toward the eradication of metastatic tumor cells must involve targeting both the tumor9s primary site of origin and those seeding in secondary tissues where metastasis has occurred. Lymph nodes represent an area of interest for targeting metastatic tumors because they play an essential role in tumor survival in non-native tissues. Interrogation of early cellular events in the draining lymph nodes (DLNs) during the initial priming of the adaptive T cell response can reveal new insights into how LN cells respond to a metastasizing tumor. The inflammatory chemokine CCL3 is important in orchestrating cellular contacts in vaccinated lymph nodes (LNs) and enhancing memory T cell generation. CCL3 has also been implicated in the modification and recruitment of natural killer (NK) cells and dendritic cells (DCs) to sites of epithelial insult and are important in establishing whether tumors will be tolerated or rejected. We hypothesize that by introducing a continuous supply of CCL3 into the microenvironment of a metastatic tumor, we can redirect a lymph node destined for tumor-tolerance toward the production of greater antitumor cellular responses. To interrogate our hypothesis, we subcutaneously inoculated naive murine recipients with a Balb/c colon metastatic tumor (CT26) that is either the wild-type (WTTUs) or WTTUs transfected to secrete CCL3 (L3TUs). Immunocompetent mice injected with L3TUs resulted in a suppression of tumor growth compared to the WTTU group in a CD8 + T cell dependent manner. 1, 3, and 5-days after injection with WTTUs or L3TUs, there was an enhanced accumulation of endogenous DCs, NKs, and T cells in the DLNs and non-draining LNs (NDLNs). In vivo analysis of DCs in the DLN of L3TUs showed increased numbers of CD11c + cells that upregulated the T cell costimulatory molecule, CD86 + , while in vitro , SIINFEKL-pulsed BMDCs cultured with CCL3, showed an enhance capacity to induce proliferation of OT-I (CD8 + ) T cells. Examination of the day-5 DLN for direct signs of adaptive antitumor responses revealed an enhanced production of the antitumor cytokine, IFNγ, in the L3TU group, while the WTTU group showed a greater accumulation of CD4 + T-regulatory cells (Treg) over convention CD4 + T cells. Together these data suggest that CCL3 may enhance the overall immune response in the DLN in three ways. First, CCL3 attracts key cell-types such as NK cells to the DLN in larger quantities that can directly interact with DCs or T cells to enhance the development of IFNγ producing immune cells. Second, CCL3 directly influences DC maturation and indirectly enhances T cell proliferation. Third, CCL3 directly enhances the overall systemic accumulation of lymphocyte and myeloid cells in the DLN and NDLNs. Citation Format: Frederick Allen, Joseph M. Nthale, Saada K. Eid, Peter Rauhe, David Askew, Jay Myers, Alexander Tong, Alex Y. Huang. CCL3 in the tumor microenvironment augments antitumor immune priming in the lymph node [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A119.


Microscopy and Microanalysis | 2011

Establishing a Novel In Vivo Platform to Interrogate the Effects of Pharmacologic and Molecularly Targeted Modulation of CDK5 in the Invasive and Metastatic Potential of Medulloblastoma in Live Experimental Animals

Agne Petrosiute; Jay Myers; Joseph Nthale; K Davis; Alex Y. Huang

Medulloblastoma (MB) is the most common malignant brain tumor of childhood. Originating from embryonal neuroepithelial cells, MB accounts for approximately 20% of all primary pediatric CNS tumors. MB often exhibits an aggressive growth pattern and is one of only a few tumor types with a metastatic potential. It can attain several centimeters in size resulting in mass-occupying lesions most often located in the posterior fossa. Due to invasion into regional subarachnoid and ventricular spaces, widespread seeding of the subarachnoid space may occur. Unfortunately, effective therapy for metastatic MB continues to be a great clinical challenge; therefore, insights into the mechanisms by which MB attains its invasive properties may spark future development of rational new therapeutic strategies.


Journal of Immunology | 2014

LFA-1 and CD62L dependent transient surface CCR5 expression in naïve CD8+ T cells enhances T cell immune memory (CCR3P.214)

Alex Y. Huang; David Askew; Charles A. Su; Rachel Liou; Joseph Nthale; Deborah Barkauskas

Collaboration


Dive into the Joseph Nthale's collaboration.

Top Co-Authors

Avatar

Alex Y. Huang

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Jay Myers

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Deborah Barkauskas

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Agne Petrosiute

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

David Askew

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Duncan Stearns

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

John J. Letterio

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Peter Rauhe

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Rodney Dixon Dorand

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Tej K. Pareek

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge