Network


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

Hotspot


Dive into the research topics where John Luff is active.

Publication


Featured researches published by John Luff.


The Journal of Neuroscience | 2003

Oxidative Stress Is Responsible for Deficient Survival and Dendritogenesis in Purkinje Neurons from Ataxia-Telangiectasia Mutated Mutant Mice

Philip Chen; Cheng Peng; John Luff; Kevin Spring; Dianne Watters; Steven E. Bottle; Shigeki Furuya; Martin F. Lavin

Atm gene-disrupted mice recapitulate the majority of characteristics observed in patients with the genetic disorder ataxia-telangiectasia (A-T). However, although they exhibit defects in neuromotor function and a distinct neurological phenotype, they do not show the progressive neurodegeneration seen in human patients, but there is evidence that ataxia-telangiectasia mutated (Atm)-deficient animals have elevated levels of oxidized macromolecules and some neuropathology. We report here that in vitro survival of cerebellar Purkinje cells from both Atm “knock-out” and Atm “knock-in” mice was significantly reduced compared with their wild-type littermates. Although most of the Purkinje neurons from wild-type mice exhibited extensive dendritic elongation and branching under these conditions, most neurons from Atm-deficient mice had dramatically reduced dendritic branching. An antioxidant (isoindoline nitroxide) prevented Purkinje cell death in Atm-deficient mice and enhanced dendritogenesis to wild-type levels. Furthermore, administration of the antioxidant throughout pregnancy had a small enhancing effect on Purkinje neuron survival in Atm gene-disrupted animals and protected against oxidative stress in older animals. These data provide strong evidence for a defect in the cerebellum of Atm-deficient mice and suggest that oxidative stress contributes to this phenotype.


PLOS Genetics | 2013

Senataxin plays an essential role with DNA damage response proteins in meiotic recombination and gene silencing.

Olivier J. Becherel; Abrey J. Yeo; Alissa Stellati; Evelyn Y. H. Heng; John Luff; Amila Suraweera; Rick Woods; Jean S Fleming; Dianne Carrie; Kristine McKinney; Xiaoling Xu; Chuxia Deng; Martin F. Lavin

Senataxin, mutated in the human genetic disorder ataxia with oculomotor apraxia type 2 (AOA2), plays an important role in maintaining genome integrity by coordination of transcription, DNA replication, and the DNA damage response. We demonstrate that senataxin is essential for spermatogenesis and that it functions at two stages in meiosis during crossing-over in homologous recombination and in meiotic sex chromosome inactivation (MSCI). Disruption of the Setx gene caused persistence of DNA double-strand breaks, a defect in disassembly of Rad51 filaments, accumulation of DNA:RNA hybrids (R-loops), and ultimately a failure of crossing-over. Senataxin localised to the XY body in a Brca1-dependent manner, and in its absence there was incomplete localisation of DNA damage response proteins to the XY chromosomes and ATR was retained on the axial elements of these chromosomes, failing to diffuse out into chromatin. Furthermore persistence of RNA polymerase II activity, altered ubH2A distribution, and abnormal XY-linked gene expression in Setx−/− revealed an essential role for senataxin in MSCI. These data support key roles for senataxin in coordinating meiotic crossing-over with transcription and in gene silencing to protect the integrity of the genome.


Journal of Clinical Neuroscience | 2008

A novel rat model for glioblastoma multiforme using a bioluminescent F98 cell line.

M.J. Bryant; Teong Chuah; John Luff; Martin F. Lavin; David G. Walker

In experimental neuro-oncology there remains a need for animal models that can be used to assess the efficacy of new and innovative treatment methodologies for glioblastoma multiforme (GBM). Rat models have remained the mainstay of neuro-oncology research for over 30 years; however, despite extensive experimentation, there is no one rat model that truly reflects the features of human tumours. We have developed a novel rat brain tumour model that closely resembles human GBM in biological behaviour and that utilizes bioluminescence imaging (BLI) to follow day-to-day in vivo progress of the tumour. F98 glioma cells were transfected with the firefly luciferase gene and injected orthotopically into the brains of 24 rats. Weekly BLI after subcutaneous injection of luciferin allowed for in vivo monitoring of the progress of the brain tumours. Euthanasia and histological analysis of the rodent brains at varying stages post-implantation, allowed for statistically significant correlation between tumour size and luminescence (p=0.002). The utility of this model is readily apparent, allowing us a way of examining the effects of new and novel therapeutics in these rats.


PLOS ONE | 2014

R-Loops in Proliferating Cells but Not in the Brain: Implications for AOA2 and Other Autosomal Recessive Ataxias

Abrey J. Yeo; Olivier J. Becherel; John Luff; Jason K. Cullen; Thidathip Wongsurawat; Piroon Jenjaroenpoon; Vladimir A. Kuznetsov; Peter J. McKinnon; Martin F. Lavin

Disruption of the Setx gene, defective in ataxia oculomotor apraxia type 2 (AOA2) leads to the accumulation of DNA/RNA hybrids (R-loops), failure of meiotic recombination and infertility in mice. We report here the presence of R-loops in the testes from other autosomal recessive ataxia mouse models, which correlate with fertility in these disorders. R-loops were coincident in cells showing high basal levels of DNA double strand breaks and in those cells undergoing apoptosis. Depletion of Setx led to high basal levels of R-loops and these were enhanced further by DNA damage both in vitro and in vivo in tissues with proliferating cells. There was no evidence for accumulation of R-loops in the brains of mice where Setx, Atm, Tdp1 or Aptx genes were disrupted. These data provide further evidence for genome destabilization as a consequence of disrupted transcription in the presence of DNA double strand breaks arising during DNA replication or recombination. They also suggest that R-loop accumulation does not contribute to the neurodegenerative phenotype in these autosomal recessive ataxias.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Smg1 haploinsufficiency predisposes to tumor formation and inflammation

Tara L. Roberts; Uda Ho; John Luff; C. Soon Lee; Simon H. Apte; Kelli P. A. MacDonald; Liza J. Raggat; Allison R. Pettit; Carl A. Morrow; Michael J. Waters; Phil Chen; Rick Woods; Gethin P. Thomas; Liam St. Pierre; Camile S. Farah; Raymond A. Clarke; James A. L. Brown; Martin F. Lavin

SMG1 is a member of the phosphoinositide kinase-like kinase family of proteins that includes ATM, ATR, and DNA-PK, proteins with known roles in DNA damage and cellular stress responses. SMG1 has a well-characterized role in nonsense-mediated decay as well as suggested roles in the DNA damage response, resistance to oxidative stress, regulation of hypoxic responses, and apoptosis. To understand the roles of SMG1 further, we generated a Genetrap Smg1 mouse model. Smg1 homozygous KO mice were early embryonic lethal, but Smg1 heterozygous mice showed a predisposition to a range of cancers, particularly lung and hematopoietic malignancies, as well as development of chronic inflammation. These mice did not display deficiencies in known roles of SMG1, including nonsense-mediated decay. However, they showed elevated basal tissue and serum cytokine levels, indicating low-level inflammation before the development of tumors. Smg1 heterozygous mice also showed evidence of oxidative damage in tissues. These data suggest that the inflammation observed in Smg1 haploinsufficiency contributes to susceptibility to cancer and that Smg1-deficient animals represent a model of inflammation-enhanced cancer development.


Genes, Chromosomes and Cancer | 2006

Regulation of the Atm promoter in vivo.

Nuri Gueven; Toshiyuki Fukao; John Luff; Carol Paterson; Graham F. Kay; Naomi Kondo; Martin F. Lavin

While ATM, the protein defective in the human genetic disorder ataxia‐telangiectasia (A‐T), is primarily activated as a preexisting protein by radiation, there is also evidence that expression of the protein can be regulated at the transcriptional level. Activation of the ATM promoter by ionizing radiation has been reported only in quiescent cells in culture. To investigate how the Atm promoter is regulated in vivo, we generated transgenic mice that express the luciferase reporter gene under the control of the murine Atm promoter. Using a biophotonic imaging system luciferase activity was monitored in vivo. Strong promoter activity was detected throughout the transgenic animals with particularly high signals from the thymus, abdominal region, and reproductive organs. This activity further increased in response to both ionizing radiation and heat stress in a time dependent manner. Luciferase activity, measured in vitro in extracts from different tissues, showed highest activities in testes, ovaries, and cerebellum. Subjecting these mice to a single dose of 4 Gy total body radiation led to a time‐dependent activation of the promoter with the strongest response observed in the peritoneal membrane, skin, and spleen. For most tissues tested, maximal promoter activity was reached 8 hr after radiation. The observed changes in promoter activity largely correlated with levels and activity of Atm protein in tissue extracts. These results demonstrate that, in addition to activation by autophosphorylation, Atm can also be regulated in vivo at the transcriptional level possibly ensuring a more sustained response to radiation and other stimuli.


Human Molecular Genetics | 2016

A rat model of ataxia-telangiectasia: evidence for a neurodegenerative phenotype

Hazel Quek; John Luff; Ka Geen Cheung; Sergei Kozlov; Magtouf Gatei; C. Soon Lee; Mark C. Bellingham; Peter G. Noakes; Yi Chieh Lim; Nigel L. Barnett; Steven Dingwall; Ernst J. Wolvetang; Tomoji Mashimo; Tara L. Roberts; Martin F. Lavin

Ataxia-telangiectasia (A-T), an autosomal recessive disease caused by mutations in the ATM gene is characterised by cerebellar atrophy and progressive neurodegeneration which has been poorly recapitulated in Atm mutant mice. Consequently, pathways leading to neurodegeneration in A-T are poorly understood. We describe here the generation of an Atm knockout rat model that does not display cerebellar atrophy but instead paralysis and spinal cord atrophy, reminiscent of that seen in older patients and milder forms of the disorder. Loss of Atm in neurons and glia leads to accumulation of cytosolic DNA, increased cytokine production and constitutive activation of microglia consistent with a neuroinflammatory phenotype. Rats lacking ATM had significant loss of motor neurons and microgliosis in the spinal cord, consistent with onset of paralysis. Since short term treatment with steroids has been shown to improve the neurological signs in A-T patients we determined if that was also the case for Atm-deficient rats. Betamethasone treatment extended the lifespan of Atm knockout rats, prevented microglial activation and significantly decreased neuroinflammatory changes and motor neuron loss. These results point to unrepaired damage to DNA leading to significant levels of cytosolic DNA in Atm-deficient neurons and microglia and as a consequence activation of the cGAS-STING pathway and cytokine production. This in turn would increase the inflammatory microenvironment leading to dysfunction and death of neurons. Thus the rat model represents a suitable one for studying neurodegeneration in A-T and adds support for the use of anti-inflammatory drugs for the treatment of neurodegeneration in A-T patients.


Journal of Immunology | 2011

Myeloma-Induced Alloreactive T Cells Arising in Myeloma-Infiltrated Bones Include Double-Positive CD8+CD4+ T Cells: Evidence from Myeloma-Bearing Mouse Model

Lisa M. Freeman; Alfred King-Yin Lam; Eugen Bogdan Petcu; Robert A. Smith; Ali Salajegheh; Peter Diamond; Andrew C.W. Zannettino; Andreas Evdokiou; John Luff; Pooi-Fong Wong; Dalia Khalil; Nigel J. Waterhouse; Frank Vari; Alison M. Rice; Laurence Catley; Derek N. J. Hart; Slavica Vuckovic

The graft-versus-myeloma (GVM) effect represents a powerful form of immune attack exerted by alloreactive T cells against multiple myeloma cells, which leads to clinical responses in multiple myeloma transplant recipients. Whether myeloma cells are themselves able to induce alloreactive T cells capable of the GVM effect is not defined. Using adoptive transfer of T naive cells into myeloma-bearing mice (established by transplantation of human RPMI8226-TGL myeloma cells into CD122+ cell-depleted NOD/SCID hosts), we found that myeloma cells induced alloreactive T cells that suppressed myeloma growth and prolonged survival of T cell recipients. Myeloma-induced alloreactive T cells arising in the myeloma-infiltrated bones exerted cytotoxic activity against resident myeloma cells, but limited activity against control myeloma cells obtained from myeloma-bearing mice that did not receive T naive cells. These myeloma-induced alloreactive T cells were derived through multiple CD8+ T cell divisions and enriched in double-positive (DP) T cells coexpressing the CD8αα and CD4 coreceptors. MHC class I expression on myeloma cells and contact with T cells were required for CD8+ T cell divisions and DP-T cell development. DP-T cells present in myeloma-infiltrated bones contained a higher proportion of cells expressing cytotoxic mediators IFN-γ and/or perforin compared with single-positive CD8+ T cells, acquired the capacity to degranulate as measured by CD107 expression, and contributed to an elevated perforin level seen in the myeloma-infiltrated bones. These observations suggest that myeloma-induced alloreactive T cells arising in myeloma-infiltrated bones are enriched with DP-T cells equipped with cytotoxic effector functions that are likely to be involved in the GVM effect.


Journal of Leukocyte Biology | 2017

Rats with a missense mutation in Atm display neuroinflammation and neurodegeneration subsequent to accumulation of cytosolic DNA following unrepaired DNA damage

Hazel Quek; John Luff; KaGeen Cheung; Sergei Kozlov; Magtouf Gatei; C. Soon Lee; Mark C. Bellingham; Peter G. Noakes; Yi Chieh Lim; Nigel L. Barnett; Steven Dingwall; Ernst J. Wolvetang; Tomoji Mashimo; Tara L. Roberts; Martin F. Lavin

Mutations in the ataxia‐telangiectasia (A‐T)‐mutated (ATM) gene give rise to the human genetic disorder A‐T, characterized by immunodeficiency, cancer predisposition, and neurodegeneration. Whereas a series of animal models recapitulate much of the A‐T phenotype, they fail to present with ataxia or neurodegeneration. We describe here the generation of an Atm missense mutant [amino acid change of leucine (L) to proline (P) at position 2262 (L2262P)] rat by intracytoplasmic injection (ICSI) of mutant sperm into oocytes. Atm‐mutant rats (AtmL2262P/L2262P) expressed low levels of ATM protein, suggesting a destabilizing effect of the mutation, and had a significantly reduced lifespan compared with Atm+/+. Whereas these rats did not show cerebellar atrophy, they succumbed to hind‐limb paralysis (45%), and the remainder developed tumors. Closer examination revealed the presence of both dsDNA and ssDNA in the cytoplasm of cells in the hippocampus, cerebellum, and spinal cord of AtmL2262P/L2262P rats. Significantly increased levels of IFN‐β and IL‐1β in all 3 tissues were indicative of DNA damage induction of the type 1 IFN response. This was further supported by NF‐κB activation, as evidenced by p65 phosphorylation (P65) and translocation to the nucleus in the spinal cord and parahippocampus. Other evidence of neuroinflammation in the brain and spinal cord was the loss of motor neurons and the presence of increased activation of microglia. These data provide support for a proinflammatory phenotype that is manifested in the Atm mutant rat as hind‐limb paralysis. This mutant represents a useful model to investigate the importance of neuroinflammation in A‐T.


Leukemia | 2015

ATM and SMG1 are tumour suppressors which co- regulate blood cancer development and cellular responses to DNA damage and oxidative stress

Patricia Reberio; Alexander James; Silvia Ling; C. Soon Lee; Uda Ho; John Luff; Hazel Quek; Martin F. Lavin; Tara L. Roberts

No. 1; Abstract Category: Cellular Biology Abstract Title: Combining CD23 chimeric antigen receptor immunotherapy and lenalidomide as a novel therapeutic strategy for chronic lymphocytic leukemia Full Name: Sarah Tettamanti Institution/Company: Centro Ricerca Tettamanti, Clinica Pediatrica, Università Milano Bicocca, Osp. San Gerardo/Fondazione MBBM, Monza, Italy; Unit of Lymphoid Malignancies, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Unit of B Cell Neoplasia, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Other Authors: Giovanni Galletti, Greta Maria Paola Giordano Attianese, Silvia Arcangeli, Tania Veliz Rodriguez, Chiara Francesca Magnani, Federica Barbaglio, Lydia Scarfò, Maurilio Ponzoni, Andrea Biondi, Federico Caligaris-Cappio, Ettore Biagi*, Paolo Ghia, Maria Teresa Sabrina Bertilaccio Maria Teresa Sabrina Bertilaccio and Paolo Ghia equally contributed to the work *Corresponding author Abstract Details: Chronic Lymphocytic Leukemia (CLL)Details: Chronic Lymphocytic Leukemia (CLL) is a chronic lymphoid malignancy characterized by immune dysfunction that particularly involves the T-cell compartment. Given the well known role of lenalidomide in enhancing T cell function [1] we exploited a novel therapeutic strategy by combining adoptive CD23.CAR-based immunotherapy with lenalidomide. First we used the CLL-xenograft model based on the injection of MEC1 CLL cell line into Rag2 / c / mice [2] to verify the in vivo antitumor activity of lenalidomide on human CLL cells. We then generated CD23.CAR T lymphocytes from 3 different peripheral blood samples collected from CD23 CLL patients, as already described for healthy donors [3]. We demonstrated that in pre-clinical settings, lenalidomide can be efficiently associated to CAR-based immunotherapy. In MEC-1 xeno-transplanted Rag2 / c / mice, by combining CAR.CD23 T cells from CLL patients with low dose lenalidomide, ineffective in monotherapy, we induced a decrease of the percentage of CD19 leukemic cells in all lymphoid and non-lymphoid tissues and an improved survival. The combination with low dose lenalidomide was more effective also when compared to human recombinant IL-2 utilized in traditional immunotherapeutic settings. In accordance with the in vivo efficacy, CAR T cells were observed in all leukemic sites suggesting an ability to migrate and home in vivo and, when purified from the bone marrow, CD23. CAR T cells were still able to mount a tumor-specific cytotoxic response in vitro. Surprisingly, CAR T cells exposed in vivo to daily lenalidomide were for the majority effector memory cells and maintained the expression of CD23. CAR on their surface. These results conceivably support the use in the CLL therapeutical setting of low doses lenalidomide to improve CAR cytotoxic response and to avoid the potential impairment of an effective immune response.

Collaboration


Dive into the John Luff's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tara L. Roberts

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Hazel Quek

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Abrey J. Yeo

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Olivier J. Becherel

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dalia Khalil

Translational Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Frank Vari

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge