Network


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

Hotspot


Dive into the research topics where Jai Rautela is active.

Publication


Featured researches published by Jai Rautela.


Nature Immunology | 2016

CIS is a potent checkpoint in NK cell–mediated tumor immunity

Rebecca B. Delconte; Tatiana B. Kolesnik; Laura F. Dagley; Jai Rautela; Wei Shi; Eva M. Putz; Kimberley Stannard; Jian Guo Zhang; Charis E. Teh; Matt Firth; Takashi Ushiki; Christopher E. Andoniou; Mariapia A. Degli-Esposti; Phillip P Sharp; C.E. Sanvitale; Giuseppe Infusini; Nicholas P. D. Liau; Edmond M. Linossi; Christopher J. Burns; Sebastian Carotta; Daniel Gray; Cyril Seillet; Dana S. Hutchinson; Gabrielle T. Belz; Andrew I. Webb; Warren S. Alexander; Shawn S.-C. Li; Alex N. Bullock; Jeffery J. Babon; Mark J. Smyth

The detection of aberrant cells by natural killer (NK) cells is controlled by the integration of signals from activating and inhibitory ligands and from cytokines such as IL-15. We identified cytokine-inducible SH2-containing protein (CIS, encoded by Cish) as a critical negative regulator of IL-15 signaling in NK cells. Cish was rapidly induced in response to IL-15, and deletion of Cish rendered NK cells hypersensitive to IL-15, as evidenced by enhanced proliferation, survival, IFN-γ production and cytotoxicity toward tumors. This was associated with increased JAK-STAT signaling in NK cells in which Cish was deleted. Correspondingly, CIS interacted with the tyrosine kinase JAK1, inhibiting its enzymatic activity and targeting JAK for proteasomal degradation. Cish−/− mice were resistant to melanoma, prostate and breast cancer metastasis in vivo, and this was intrinsic to NK cell activity. Our data uncover a potent intracellular checkpoint in NK cell–mediated tumor immunity and suggest possibilities for new cancer immunotherapies directed at blocking CIS function.


Nature Immunology | 2017

Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells

Yulong Gao; Fernando Souza-Fonseca-Guimaraes; Tobias Bald; Susanna S. Ng; Arabella Young; Shin Foong Ngiow; Jai Rautela; Jasmin Straube; Nic Waddell; Stephen J. Blake; Juming Yan; Laurent Bartholin; Jason S. Lee; Eric Vivier; Kazuyoshi Takeda; Meriem Messaoudene; Laurence Zitvogel; Michele W.L. Teng; Gabrielle T. Belz; Christian R. Engwerda; Nicholas D. Huntington; Kyohei Nakamura; Michael Hölzel; Mark J. Smyth

Avoiding destruction by immune cells is a hallmark of cancer, yet how tumors ultimately evade control by natural killer (NK) cells remains incompletely defined. Using global transcriptomic and flow-cytometry analyses and genetically engineered mouse models, we identified the cytokine-TGF-β-signaling-dependent conversion of NK cells (CD49a−CD49b+Eomes+) into intermediate type 1 innate lymphoid cell (intILC1) (CD49a+CD49b+Eomes+) populations and ILC1 (CD49a+CD49b−Eomesint) populations in the tumor microenvironment. Strikingly, intILC1s and ILC1s were unable to control local tumor growth and metastasis, whereas NK cells favored tumor immunosurveillance. Experiments with an antibody that neutralizes the cytokine TNF suggested that escape from the innate immune system was partially mediated by TNF-producing ILC1s. Our findings provide new insight into the plasticity of group 1 ILCs in the tumor microenvironment and suggest that the TGF-β-driven conversion of NK cells into ILC1s is a previously unknown mechanism by which tumors escape surveillance by the innate immune system.


Cancer Research | 2014

BMP4 Inhibits Breast Cancer Metastasis by Blocking Myeloid-Derived Suppressor Cell Activity

Yuan Cao; Clare Y. Slaney; Bradley N. Bidwell; Belinda S. Parker; Cameron N. Johnstone; Jai Rautela; Bedrich L. Eckhardt; Robin L. Anderson

The TGFβ growth factor family member BMP4 is a potent suppressor of breast cancer metastasis. In the mouse, the development of highly metastatic mammary tumors is associated with an accumulation of myeloid-derived suppressor cells (MDSC), the numbers of which are reduced by exogenous BMP4 expression. MDSCs are undetectable in naïve mice but can be induced by treatment with granulocyte colony-stimulating factor (G-CSF/Csf3) or by secretion of G-CSF from the tumor. Both tumor-induced and G-CSF-induced MDSCs effectively suppress T-cell activation and proliferation, leading to metastatic enhancement. BMP4 reduces the expression and secretion of G-CSF by inhibiting NF-κB (Nfkb1) activity in human and mouse tumor lines. Because MDSCs correlate with poor prognosis in patients with breast cancer, therapies based on activation of BMP4 signaling may offer a novel treatment strategy for breast cancer. Cancer Res; 74(18); 5091-102. ©2014 AACR.


Cancer Research | 2013

The Emerging Role of Immunosurveillance in Dictating Metastatic Spread in Breast Cancer

Clare Y. Slaney; Jai Rautela; Belinda S. Parker

It is now well known that the immune system can recognize transformed cells and control the initiation and growth of some cancers, a process termed tumor immunosurveillance. Key regulators of this process have been described in the primary tumor setting, where the balance of protumor and antitumor responses dictates tumor initiation and progression. Accumulating evidence suggests that immunosurveillance may also be critical for regulating metastatic spread, the most fatal aspect of cancer, and that mechanisms of overcoming immune control may be quite different from those at the primary site. Our recent findings support loss of type I interferon (IFN) signaling as a tumor-cell intrinsic mechanism of evading metastasis-specific immune responses in breast cancer. We revealed that type I IFN-induced innate (natural killer) and adaptive (CD8(+) T cell) responses suppressed bone metastatic growth and this was associated with decreased accumulation of immune suppressor cells (myeloid-derived suppressor cells). This review summarizes recent findings that are in support of tumor-induced immunosurveillance in regulating metastatic spread, including evidence that immune regulation of primary tumors may be distinct from those dictating metastasis.


Cancer immunology research | 2015

Loss of Host Type-I IFN Signaling Accelerates Metastasis and Impairs NK-cell Antitumor Function in Multiple Models of Breast Cancer

Jai Rautela; Nikola Baschuk; Clare Y. Slaney; Krishnath M. Jayatilleke; Kun Xiao; Bradley N. Bidwell; Erin Lucas; Edwin D. Hawkins; Peter Lock; Christina S.F. Wong; Weisan Chen; Robin L. Anderson; Paul J. Hertzog; Daniel M. Andrews; Andreas Möller; Belinda S. Parker

Type-I interferon immune signaling plays a critical role during the antimetastatic immune response. The authors show that loss of this pathway can promote bone metastasis in three different breast cancer models, revealing its importance across different models. Metastatic progression is the major cause of breast cancer–related mortality. By examining multiple syngeneic preclinical breast cancer models in mice lacking a functional type-I interferon receptor (Ifnar1−/− mice), we show that host-derived type-I interferon (IFN) signaling is a critical determinant of metastatic spread that is independent of primary tumor growth. In particular, we show that bone metastasis can be accelerated in Balb/c Ifnar1−/− mice bearing either 4T1 or 66cl4 orthotopic tumors and, for the first time, present data showing the development of bone metastasis in the C57Bl/6 spontaneous MMTV-PyMT–driven model of tumorigenesis. Further exploration of these results revealed that endogenous type-I IFN signaling to the host hematopoietic system is a key determinant of metastasis-free survival and critical to the responsiveness of the circulating natural killer (NK)–cell population. We find that in vivo–stimulated NK cells derived from wild-type, but not Ifnar1−/−, mice can eliminate the 4T1 and 66cl4 breast tumor lines with varying kinetics in vitro. Together, this study indicates that the dysregulated immunity resulting from a loss of host type-I IFN signaling is sufficient to drive metastasis, and provides a rationale for targeting the endogenous type-I IFN pathway as an antimetastatic strategy. Cancer Immunol Res; 3(11); 1207–17. ©2015 AACR.


Current Opinion in Immunology | 2017

IL-15 signaling in NK cell cancer immunotherapy.

Jai Rautela; Nicholas D. Huntington

While cancer has been traditionally treated by chemotherapy, radiation, targeted therapies and surgery, a fifth pillar of cancer treatment, immunotherapy, has emerged over the past 10 years and revolutionized our war on cancer. The benchmark for drugs in this category has been set by the development of CD8 T cell checkpoint (CTLA-4 and PD-1/PD-L1) inhibitors. These therapies effectively expand and reactivate the pool of tumor-specific T cells leading to objective response rates of up to 50% in patients with certain cancers. However, the significant number of patients and cancer types that altogether fail or acquire resistance to these therapies highlights the need for novel immunotherapies that target alternate pathways and effector cells. Thus, there is renewed interest in harnessing the tumor-killing abilities of Natural Killer (NK) cells, though it has proven difficult to efficiently and specifically target these cells cancer patients. The commercial success of T cell checkpoint inhibitors has seen a swam of new biotech companies emerge with innovative or revised strategies that aim to harness the innate non-antigen dependent tumor lysis potential of NK cells. This review will focus on IL-15 biology in NK cells and proposes the development novel therapies aimed at this pathway in humans.


Oncotarget | 2015

Cysteine cathepsin activity suppresses osteoclastogenesis of myeloid-derived suppressor cells in breast cancer

Laura E. Edgington-Mitchell; Jai Rautela; Hendrika M. Duivenvoorden; Krishnath M. Jayatilleke; Wouter A. van der Linden; Martijn Verdoes; Matthew Bogyo; Belinda S. Parker

Cysteine cathepsin proteases contribute to many normal cellular functions, and their aberrant activity within various cell types can contribute to many diseases, including breast cancer. It is now well accepted that cathepsin proteases have numerous cell-specific functions within the tumor microenvironment that function to promote tumor growth and invasion, such that they may be valid targets for anti-metastatic therapeutic approaches. Using activity-based probes, we have examined the activity and expression of cysteine cathepsins in a mouse model of breast cancer metastasis to bone. In mice bearing highly metastatic tumors, we detected abundant cysteine cathepsin expression and activity in myeloid-derived suppressor cells (MDSCs). These immature immune cells have known metastasis-promoting roles, including immunosuppression and osteoclastogenesis, and we assessed the contribution of cysteine cathepsins to these functions. Blocking cysteine cathepsin activity with multiple small-molecule inhibitors resulted in enhanced differentiation of multinucleated osteoclasts. This highlights a potential role for cysteine cathepsin activity in suppressing the fusion of osteoclast precursor cells. In support of this hypothesis, we found that expression and activity of key cysteine cathepsins were downregulated during MDSC-osteoclast differentiation. Another cysteine protease, legumain, also inhibits osteoclastogenesis, in part through modulation of cathepsin L activity. Together, these data suggest that cysteine protease inhibition is associated with enhanced osteoclastogenesis, a process that has been implicated in bone metastasis.


Cancer Research | 2017

Targeting Adenosine in BRAF-Mutant Melanoma Reduces Tumor Growth and Metastasis

Arabella Young; Shin Foong Ngiow; Jason Madore; Julia Reinhardt; Jennifer Landsberg; Arash Chitsazan; Jai Rautela; Tobias Bald; Deborah S. Barkauskas; Elizabeth Ahern; Nicholas D. Huntington; Dirk Schadendorf; Glen M. Boyle; Michael Hölzel; Richard A. Scolyer; Mark J. Smyth

Increasing evidence exists for the role of immunosuppressive adenosine in promoting tumor growth and spread in a number of cancer types, resulting in poor clinical outcomes. In this study, we assessed whether the CD73-adenosinergic pathway is active in melanoma patients and whether adenosine restricts the efficacy of clinically approved targeted therapies for commonly mutated BRAFV600E melanoma. In AJCC stage III melanoma patients, CD73 expression (the enzyme that generates adenosine) correlated significantly with patients presenting nodal metastatic melanoma, suggesting that targeting this pathway may be effective in advanced stage disease. In addition, dabrafenib and trametinib treatment of CD73+ BRAFV600E-mutant melanomas caused profound CD73 downregulation in tumor cells. Inhibition of BRAF and MEK in combination with the A2A adenosine receptor provided significant protection against tumor initiation and metastasis formation in mice. Our results suggest that targeting adenosine may enhance therapeutic responses for melanoma patients receiving targeted or immune-based therapies. Cancer Res; 77(17); 4684-96. ©2017 AACR.


Journal of Experimental Medicine | 2017

Cell cycle progression dictates the requirement for BCL2 in natural killer cell survival

Charlotte Viant; Sophie Guia; Robert J. Hennessy; Jai Rautela; Kim Pham; Claire Bernat; Wilford Goh; Yuhao Jiao; Rebecca B. Delconte; Michael Roger; Vanina Simon; Fernando Souza-Fonseca-Guimaraes; Stephanie Grabow; Gabrielle T. Belz; Benjamin T. Kile; Andreas Strasser; Daniel Gray; Phillip D. Hodgkin; Bruce Beutler; Eric Vivier; Sophie Ugolini; Nicholas D. Huntington

Natural killer (NK) cells are innate lymphoid cells with antitumor functions. Using an N-ethyl-N-nitrosourea (ENU)–induced mutagenesis screen in mice, we identified a strain with an NK cell deficiency caused by a hypomorphic mutation in the Bcl2 (B cell lymphoma 2) gene. Analysis of these mice and the conditional deletion of Bcl2 in NK cells revealed a nonredundant intrinsic requirement for BCL2 in NK cell survival. In these mice, NK cells in cycle were protected against apoptosis, and NK cell counts were restored in inflammatory conditions, suggesting a redundant role for BCL2 in proliferating NK cells. Consistent with this, cycling NK cells expressed higher MCL1 (myeloid cell leukemia 1) levels in both control and BCL2-null mice. Finally, we showed that deletion of BIM restored survival in BCL2-deficient but not MCL1-deficient NK cells. Overall, these data demonstrate an essential role for the binding of BCL2 to BIM in the survival of noncycling NK cells. They also favor a model in which MCL1 is the dominant survival protein in proliferating NK cells.


Immunology and Cell Biology | 2018

Molecular insight into targeting the NK cell immune response to cancer

Jai Rautela; Fernando Souza-Fonseca-Guimaraes; Soroor Hediyeh-Zadeh; Rebecca B. Delconte; Melissa J. Davis; Nicholas D. Huntington

Natural Killer (NK) cells have long been considered an important part of the anti‐tumor immune response due to their potent cytolytic and cytokine‐secreting abilities. To date, a clear demonstration of the role NK cells play in human cancer is lacking, and there are still very few examples of therapies that efficiently exploit or enhance the spontaneous ability of NK cells to destroy the autologous cancer cells. Given the paradigm shift toward cancer immunotherapy over the past decade, there is a renewed push to understand how NK cell homeostasis and function are regulated in order to therapeutically harness these cells to treat cancer. This review will highlight recent advancements in our understanding of how growth factors impact on NK cell development, differentiation, survival and function with an emphasis on how these pathways may influence NK cell activity in the tumor microenvironment and control of cancer metastasis.

Collaboration


Dive into the Jai Rautela's collaboration.

Top Co-Authors

Avatar

Nicholas D. Huntington

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rebecca B. Delconte

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bradley N. Bidwell

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Gabrielle T. Belz

Walter and Eliza Hall Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark J. Smyth

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge