55 results on '"Jacob T. Jackson"'
Search Results
2. The Haematopoietically-expressed homeobox transcription factor: roles in development, physiology and disease
- Author
-
Jacob T. Jackson, Stephen L. Nutt, and Matthew P. McCormack
- Subjects
transcription factor ,Hhex ,diabetes ,haematopoiesis ,leakamia ,AML ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The Haematopoietically expressed homeobox transcription factor (Hhex) is a transcriptional repressor that is of fundamental importance across species, as evident by its evolutionary conservation spanning fish, amphibians, birds, mice and humans. Indeed, Hhex maintains its vital functions throughout the lifespan of the organism, beginning in the oocyte, through fundamental stages of embryogenesis in the foregut endoderm. The endodermal development driven by Hhex gives rise to endocrine organs such as the pancreas in a process which is likely linked to its role as a risk factor in diabetes and pancreatic disorders. Hhex is also required for the normal development of the bile duct and liver, the latter also importantly being the initial site of haematopoiesis. These haematopoietic origins are governed by Hhex, leading to its crucial later roles in definitive haematopoietic stem cell (HSC) self-renewal, lymphopoiesis and haematological malignancy. Hhex is also necessary for the developing forebrain and thyroid gland, with this reliance on Hhex evident in its role in endocrine disorders later in life including a potential role in Alzheimer’s disease. Thus, the roles of Hhex in embryological development throughout evolution appear to be linked to its later roles in a variety of disease processes.
- Published
- 2023
- Full Text
- View/download PDF
3. Hhex Directly Represses BIM-Dependent Apoptosis to Promote NK Cell Development and Maintenance
- Author
-
Wilford Goh, Sebastian Scheer, Jacob T. Jackson, Soroor Hediyeh-Zadeh, Rebecca B. Delconte, Iona S. Schuster, Christopher E. Andoniou, Jai Rautela, Mariapia A. Degli-Esposti, Melissa J. Davis, Matthew P. McCormack, Stephen L. Nutt, and Nicholas D. Huntington
- Subjects
NK cells ,proliferation ,survival ,apoptosis ,BIM ,transcriptional regulation ,Biology (General) ,QH301-705.5 - Abstract
Summary: Hhex encodes a homeobox transcriptional regulator important for embryonic development and hematopoiesis. Hhex is highly expressed in NK cells, and its germline deletion results in significant defects in lymphoid development, including NK cells. To determine if Hhex is intrinsically required throughout NK cell development or for NK cell function, we generate mice that specifically lack Hhex in NK cells. NK cell frequency is dramatically reduced, while NK cell differentiation, IL-15 responsiveness, and function at the cellular level remain largely normal in the absence of Hhex. Increased IL-15 availability fails to fully reverse NK lymphopenia following conditional Hhex deletion, suggesting that Hhex regulates developmental pathways extrinsic to those dependent on IL-15. Gene expression and functional genetic approaches reveal that Hhex regulates NK cell survival by directly binding Bcl2l11 (Bim) and repressing expression of this key apoptotic mediator. These data implicate Hhex as a transcriptional regulator of NK cell homeostasis and immunity.
- Published
- 2020
- Full Text
- View/download PDF
4. Hhex induces promyelocyte self-renewal and cooperates with growth factor independence to cause myeloid leukemia in mice
- Author
-
Jacob T. Jackson, Ashley P. Ng, Benjamin J. Shields, Sue Haupt, Ygal Haupt, and Matthew P. McCormack
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: The hematopoietically expressed homeobox (Hhex) transcription factor is overexpressed in human myeloid leukemias. Conditional knockout models of murine acute myeloid leukemia indicate that Hhex maintains leukemia stem cell self-renewal by enabling Polycomb-mediated epigenetic repression of the Cdkn2a tumor suppressor locus, encoding p16Ink4a and p19Arf. However, whether Hhex overexpression also affects hematopoietic differentiation is unknown. To study this, we retrovirally overexpressed Hhex in hematopoietic progenitors. This enabled serial replating of myeloid progenitors, leading to the rapid establishment of interleukin-3 (IL-3)–dependent promyelocytic cell lines. Use of a Hhex-ERT2 fusion protein demonstrated that continuous nuclear Hhex is required for transformation, and structure function analysis demonstrated a requirement of the DNA-binding and N-terminal–repressive domains of Hhex for promyelocytic transformation. This included the N-terminal promyelocytic leukemia protein (Pml) interaction domain, although deletion of Pml failed to prevent Hhex-induced promyelocyte transformation, implying other critical partners. Furthermore, deletion of p16Ink4a or p19Arf did not promote promyelocyte transformation, indicating that repression of distinct Hhex target genes is required for this process. Indeed, transcriptome analysis showed that Hhex overexpression resulted in repression of several myeloid developmental genes. To test the potential for Hhex overexpression to contribute to leukemic transformation, Hhex-transformed promyelocyte lines were rendered growth factor–independent using a constitutively active IL-3 receptor common β subunit (βcV449E). The resultant cell lines resulted in a rapid promyelocytic leukemia in vivo. Thus, Hhex overexpression can contribute to myeloid leukemia via multiple mechanisms including differentiation blockade and enabling epigenetic repression of the Cdkn2a locus.
- Published
- 2018
- Full Text
- View/download PDF
5. G-CSF drives autoinflammation in APLAID
- Author
-
Elisabeth Mulazzani, Klara Kong, Juan I. Aróstegui, Ashley P. Ng, Nishika Ranathunga, Waruni Abeysekera, Alexandra L. Garnham, Sze-Ling Ng, Paul J. Baker, Jacob T. Jackson, John D. Lich, Margaret L. Hibbs, Ian P. Wicks, Cynthia Louis, and Seth L. Masters
- Subjects
Immunology ,Immunology and Allergy - Abstract
Missense mutations in PLCG2 can cause autoinflammation with phospholipase C gamma 2-associated antibody deficiency and immune dysregulation (APLAID). Here, we generated a mouse model carrying an APLAID mutation (p.Ser707Tyr) and found that inflammatory infiltrates in the skin and lungs were only partially ameliorated by removing inflammasome function via the deletion of caspase-1. Also, deleting interleukin-6 or tumor necrosis factor did not fully prevent APLAID mutant mice from autoinflammation. Overall, these findings are in accordance with the poor response individuals with APLAID have to treatments that block interleukin-1, JAK1/2 or tumor necrosis factor. Cytokine analysis revealed increased granulocyte colony-stimulating factor (G-CSF) levels as the most distinct feature in mice and individuals with APLAID. Remarkably, treatment with a G-CSF antibody completely reversed established disease in APLAID mice. Furthermore, excessive myelopoiesis was normalized and lymphocyte numbers rebounded. APLAID mice were also fully rescued by bone marrow transplantation from healthy donors, associated with reduced G-CSF production, predominantly from non-hematopoietic cells. In summary, we identify APLAID as a G-CSF-driven autoinflammatory disease, for which targeted therapy is feasible.
- Published
- 2023
- Full Text
- View/download PDF
6. Novel steganography detection using an artificial immune system approach.
- Author
-
Jacob T. Jackson, Gregg H. Gunsch, Roger L. Claypoole Jr., and Gary B. Lamont
- Published
- 2003
- Full Text
- View/download PDF
7. Wavelet-based steganalysis using a computational immune system approach.
- Author
-
Jacob T. Jackson, Gregg H. Gunsch, Roger L. Claypoole Jr., and Gary B. Lamont
- Published
- 2003
8. T-ALL can evolve to oncogene independence
- Author
-
Benjamin J. Shields, Nicholas C. Wong, David J. Curtis, Pieter Van Vlierberghe, Ross A. Dickins, Feng Yan, Matthew P. McCormack, Warren S. Alexander, Tin Wong, Tenae J. Davies, Jacob T. Jackson, Elizabeth M. Viney, Lisa Demoen, Hesham Abdulla, Anh Vo, and Raed Alserihi
- Subjects
0301 basic medicine ,LMO2 ,Cancer Research ,Oncogene ,Hematology ,Biology ,Oncogene Addiction ,medicine.disease ,Ikaros Transcription Factor ,03 medical and health sciences ,Leukemia ,030104 developmental biology ,0302 clinical medicine ,Oncology ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Stem cell ,Transcription factor ,TAL1 - Abstract
The majority of cases of T-cell acute lymphoblastic leukemia (T-ALL) contain chromosomal abnormalities that drive overexpression of oncogenic transcription factors. However, whether these initiating oncogenes are required for leukemia maintenance is poorly understood. To address this, we developed a tetracycline-regulated mouse model of T-ALL driven by the oncogenic transcription factor Lmo2. This revealed that whilst thymus-resident pre-Leukemic Stem Cells (pre-LSCs) required continuous Lmo2 expression, the majority of leukemias relapsed despite Lmo2 withdrawal. Relapse was associated with a mature phenotype and frequent mutation or loss of tumor suppressor genes including Ikzf1 (Ikaros), with targeted deletion Ikzf1 being sufficient to transform Lmo2-dependent leukemias to Lmo2-independence. Moreover, we found that the related transcription factor TAL1 was dispensable in several human T-ALL cell lines that contain SIL-TAL1 chromosomal deletions driving its overexpression, indicating that evolution to oncogene independence can also occur in human T-ALL. Together these results indicate an evolution of oncogene addiction in murine and human T-ALL and show that loss of Ikaros is a mechanism that can promote self-renewal of T-ALL lymphoblasts in the absence of an initiating oncogenic transcription factor.
- Published
- 2021
- Full Text
- View/download PDF
9. The transcription factor IRF4 represses proapoptotic BMF and BIM to licence multiple myeloma survival
- Author
-
Simon N. Willis, Charis E Teh, Yang Liao, Mark F. van Delft, Tania Tan, George Grigoriadis, Lorraine A. O'Reilly, Stephen L. Nutt, David C.S. Huang, Jia-Nan Gong, Julie Tellier, Michael S.Y. Low, Marco J Herold, Yuan Yao, Jacob T. Jackson, Wei Shi, Lin Tai, and Pasquale L. Fedele
- Subjects
Cancer Research ,medicine.medical_specialty ,Hematology ,Bcl-2 family ,Signal transducing adaptor protein ,Biology ,medicine.disease ,Oncology ,Apoptosis ,Internal medicine ,Cancer research ,medicine ,Transcription factor ,Multiple myeloma ,IRF4 ,Interferon regulatory factors - Published
- 2020
- Full Text
- View/download PDF
10. A new lymphoid-primed progenitor marked by Dach1 downregulation identified with single cell multi-omics
- Author
-
Matthew P. McCormack, Sara Tomei, Olivia Stonehouse, Adrienne A. Hilton, Matthew E. Ritchie, Andrew Jarratt, Dawn S. Lin, Warren S. Alexander, Samir Taoudi, Mark D. McKenzie, Stephen L. Nutt, Jaring Schreuder, Ashley P. Ng, Shalin H. Naik, Ladina Di Rago, Daniela Amann-Zalcenstein, Jacob T. Jackson, Jessica E. Bolden, Carolyn A. de Graaf, Kirsten A. Fairfax, and Luyi Tian
- Subjects
0301 basic medicine ,Myeloid ,Immunology ,Biology ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Single-cell analysis ,Megakaryocyte ,medicine ,Immunology and Allergy ,Lymphoid Progenitor Cells ,Lymphopoiesis ,Progenitor cell ,030215 immunology ,Progenitor - Abstract
A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.
- Published
- 2020
- Full Text
- View/download PDF
11. Protein kinase R is an innate immune sensor of proteotoxic stress via accumulation of cytoplasmic IL-24
- Author
-
Sophia Davidson, Chien-Hsiung Yu, Annemarie Steiner, Frédéric Ebstein, Paul J. Baker, Valentina Jarur-Chamy, Katja Hrovat Schaale, Pawat Laohamonthonkul, Klara Kong, Dale J. Calleja, Cassandra R. Harapas, Katherine R. Balka, Jacob Mitchell, Jacob T. Jackson, Niall D. Geoghegan, Fiona Moghaddas, Kelly L. Rogers, Katrin D. Mayer-Barber, Adriana A. De Jesus, Dominic De Nardo, Benjamin T. Kile, Anthony J. Sadler, M. Cecilia Poli, Elke Krüger, Raphaela Goldbach Mansky, and Seth L. Masters
- Subjects
Mice, Inbred C57BL ,Mice, Knockout ,Mice ,eIF-2 Kinase ,Interleukins ,Immunology ,Animals ,Humans ,General Medicine ,Cells, Cultured ,Immunity, Innate - Abstract
Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αβ) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor–induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum–associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.
- Published
- 2022
- Full Text
- View/download PDF
12. Detecting novel steganography with an anomaly-based strategy.
- Author
-
Jacob T. Jackson, Gregg H. Gunsch, Roger L. Claypoole Jr., and Gary B. Lamont
- Published
- 2004
- Full Text
- View/download PDF
13. The NUP98-HOXD13 fusion oncogene induces thymocyte self-renewal via Lmo2/Lyl1
- Author
-
Ngoc Vo, Christopher Slape, Wei Shi, Benjamin J. Shields, Jacob T. Jackson, Hansini Ranasinghe, David J. Curtis, Matthew P. McCormack, and Adriana Pliego-Zamora
- Subjects
0301 basic medicine ,LMO2 ,Cancer Research ,Oncogene Proteins, Fusion ,T cell ,Mice, Transgenic ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Mice ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Progenitor cell ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Homeodomain Proteins ,Thymocytes ,Hematology ,LIM Domain Proteins ,medicine.disease ,Neoplasm Proteins ,Mice, Inbred C57BL ,Nuclear Pore Complex Proteins ,Haematopoiesis ,Thymocyte ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Transcription Factors ,TAL1 - Abstract
T cell acute lymphoblastic leukaemia (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. While it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukaemia (AML) as well as T-ALL. We find that thymocytes from preleukaemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional programme closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression programme in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus multiple T cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1; however, NHD13 can also promote T-ALL via an alternative pathway.
- Published
- 2019
- Full Text
- View/download PDF
14. OBF1 and Oct factors control the germinal center transcriptional program
- Author
-
Michael B. Stadler, Gerhard Christofori, Fabian Wu, Chun Cao, Stefan Dirnhofer, Stephen L. Nutt, Fengyuan Tang, Mathias Frederiksen, Patrick Matthias, Hubertus Kohler, Barna D. Fodor, Simon N. Willis, Mohamed-Amin Choukrallah, Shuang Song, and Jacob T. Jackson
- Subjects
0301 basic medicine ,Lipopolysaccharides ,Chromatin Immunoprecipitation ,Transcription, Genetic ,Immunology ,Mice, Transgenic ,Biology ,Biochemistry ,Proto-Oncogene Protein c-ets-1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Coactivator ,Animals ,Humans ,RNA, Small Interfering ,Enhancer ,Transcription factor ,B-Lymphocytes ,POU domain ,Lymphoma, Non-Hodgkin ,Germinal center ,Promoter ,Cell Biology ,Hematology ,BCL6 ,Germinal Center ,Recombinant Proteins ,Cell biology ,Chromatin ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Ontology ,HEK293 Cells ,030220 oncology & carcinogenesis ,Trans-Activators ,RNA Interference ,Octamer Transcription Factor-2 ,Octamer Transcription Factor-1 - Abstract
OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell–specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.
- Published
- 2020
15. The role of PLCγ2 in immunological disorders, cancer, and neurodegeneration
- Author
-
Seth L. Masters, Jacob T. Jackson, Elisabeth Mulazzani, and Stephen L. Nutt
- Subjects
0301 basic medicine ,TB, tuberculosis ,PLAID, PLCγ2-associated antibody deficiency and immune dysregulation ,Syk ,RA, rheumatoid arthritis ,Lymphocyte proliferation ,medicine.disease_cause ,Biochemistry ,APLAID, PLCγ2-associated antibody deficiency and immune dysregulation with autoinflammation ,SLE, systemic lupus erythematosus ,Neoplasms ,BCR, B cell receptor ,Agammaglobulinaemia Tyrosine Kinase ,DLBCL, diffuse large B cell lymphoma ,phospholipase C ,Cbl, c-Casitas B-lineage lymphoma ,biology ,CLL, chronic lymphocytic leukemia ,TCR, T cell receptor ,Neurodegeneration ,neurodegeneration ,Neurodegenerative Diseases ,EBL, endemic Burkitt lymphoma ,Cell biology ,Immune System Diseases ,Second messenger system ,EBV, Epstein–Barr virus ,Signal transduction ,DCs, dendritic cells ,CVID, common variable immunodeficiency ,Aβ, amyloid β ,IP3, inositol 1,4,5-trisphosphate ,AD, Alzheimer’s disease ,cSH2, C-terminal Src homology 2 ,PLCγ2, phosphatidylinositol-specific phospholipase Cγ2 ,03 medical and health sciences ,ROS, reactive oxygen species ,RANKL, receptor activator of nuclear factor kappa-B ligand ,Calcium flux ,medicine ,Bruton's tyrosine kinase ,Animals ,Humans ,Syk Kinase ,cancer ,Calcium Signaling ,Molecular Biology ,030102 biochemistry & molecular biology ,Phospholipase C gamma ,JBC Reviews ,Cell Biology ,Immune dysregulation ,medicine.disease ,Ig, immunoglobulin ,030104 developmental biology ,inflammation ,PIP2, phosphatidylinositol-4,5-bisphosphate ,biology.protein ,BM, bone marrow ,SLP-76, SH2 domain–containing leukocyte protein of 76 kDa ,immunodeficiency ,DAG, diacylglycerol - Abstract
Phosphatidylinositol-specific phospholipase Cγ2 (PLCγ2) is a critical signaling molecule activated downstream from a variety of cell surface receptors that contain an intracellular immunoreceptor tyrosine-based activation motif. These receptors recruit kinases such as Syk, BTK, and BLNK to phosphorylate and activate PLCγ2, which then generates 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol. These well-known second messengers are required for diverse membrane functionality including cellular proliferation, endocytosis, and calcium flux. As a result, PLCγ2 dysfunction is associated with a variety of diseases including cancer, neurodegeneration, and immune disorders. The diverse pathologies associated with PLCγ2 are exemplified by distinct genetic variants. Inherited mutations at this locus cause PLCγ2-associated antibody deficiency and immune dysregulation, in some cases with autoinflammation. Acquired mutations at this locus, which often arise as a result of BTK inhibition to treat chronic lymphocytic leukemia, result in constitutive downstream signaling and lymphocyte proliferation. Finally, a third group of PLCγ2 variants actually has a protective effect in a variety of neurodegenerative disorders, presumably by increased uptake and degradation of deleterious neurological aggregates. Therefore, manipulating PLCγ2 activity either up or down could have therapeutic benefit; however, we require a better understanding of the signaling pathways propagated by these variants before such clinical utility can be realized. Here, we review the signaling roles of PLCγ2 in hematopoietic cells to help understand the effect of mutations driving immune disorders and cancer and extrapolate from this to roles which may relate to protection against neurodegeneration.
- Published
- 2020
16. Hhex Directly Represses BIM-Dependent Apoptosis to Promote NK Cell Development and Maintenance
- Author
-
Matthew P. McCormack, Stephen L. Nutt, Jacob T. Jackson, Sebastian Scheer, Wilford Goh, Nicholas D. Huntington, Christopher E. Andoniou, Rebecca B. Delconte, Soroor Hediyeh-Zadeh, Jai Rautela, Mariapia A. Degli-Esposti, Melissa J. Davis, and Iona S. Schuster
- Subjects
0301 basic medicine ,Male ,Cell Survival ,Cellular differentiation ,proliferation ,Cell ,NK cells ,Biology ,survival ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Transcriptional regulation ,Animals ,BIM ,transcriptional regulation ,Transcription factor ,lcsh:QH301-705.5 ,Cell Proliferation ,Regulation of gene expression ,Homeodomain Proteins ,Interleukin-15 ,Cell growth ,apoptosis ,Cell Differentiation ,Cell biology ,Hematopoiesis ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,BCL2L11 ,Gene Expression Regulation ,lcsh:Biology (General) ,Female ,Signal transduction ,030217 neurology & neurosurgery ,Signal Transduction ,Transcription Factors - Abstract
Summary: Hhex encodes a homeobox transcriptional regulator important for embryonic development and hematopoiesis. Hhex is highly expressed in NK cells, and its germline deletion results in significant defects in lymphoid development, including NK cells. To determine if Hhex is intrinsically required throughout NK cell development or for NK cell function, we generate mice that specifically lack Hhex in NK cells. NK cell frequency is dramatically reduced, while NK cell differentiation, IL-15 responsiveness, and function at the cellular level remain largely normal in the absence of Hhex. Increased IL-15 availability fails to fully reverse NK lymphopenia following conditional Hhex deletion, suggesting that Hhex regulates developmental pathways extrinsic to those dependent on IL-15. Gene expression and functional genetic approaches reveal that Hhex regulates NK cell survival by directly binding Bcl2l11 (Bim) and repressing expression of this key apoptotic mediator. These data implicate Hhex as a transcriptional regulator of NK cell homeostasis and immunity.
- Published
- 2020
17. T-ALL can evolve to oncogene independence
- Author
-
Hesham, Abdulla, Anh, Vo, Benjamin J, Shields, Tenae J, Davies, Jacob T, Jackson, Raed, Alserihi, Elizabeth M, Viney, Tin, Wong, Feng, Yan, Nicholas C, Wong, Lisa, Demoen, David J, Curtis, Warren S, Alexander, Pieter, Van Vlierberghe, Ross A, Dickins, and Matthew P, McCormack
- Subjects
Mice, Inbred C57BL ,Mice, Knockout ,Ikaros Transcription Factor ,Mice ,Gene Expression Regulation, Leukemic ,Animals ,Oncogenes ,LIM Domain Proteins ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Adaptor Proteins, Signal Transducing - Abstract
The majority of cases of T-cell acute lymphoblastic leukemia (T-ALL) contain chromosomal abnormalities that drive overexpression of oncogenic transcription factors. However, whether these initiating oncogenes are required for leukemia maintenance is poorly understood. To address this, we developed a tetracycline-regulated mouse model of T-ALL driven by the oncogenic transcription factor Lmo2. This revealed that whilst thymus-resident pre-Leukemic Stem Cells (pre-LSCs) required continuous Lmo2 expression, the majority of leukemias relapsed despite Lmo2 withdrawal. Relapse was associated with a mature phenotype and frequent mutation or loss of tumor suppressor genes including Ikzf1 (Ikaros), with targeted deletion Ikzf1 being sufficient to transform Lmo2-dependent leukemias to Lmo2-independence. Moreover, we found that the related transcription factor TAL1 was dispensable in several human T-ALL cell lines that contain SIL-TAL1 chromosomal deletions driving its overexpression, indicating that evolution to oncogene independence can also occur in human T-ALL. Together these results indicate an evolution of oncogene addiction in murine and human T-ALL and show that loss of Ikaros is a mechanism that can promote self-renewal of T-ALL lymphoblasts in the absence of an initiating oncogenic transcription factor.
- Published
- 2020
18. Author response for 'Hhex regulates murine lymphoid progenitor survival independently of Stat5 and Cdkn2a'
- Author
-
David M. Tarlinton, Amanda Light, Matthew P. McCormack, Wilford Goh, Jacob T. Jackson, Kristy O'Donnell, and Nicholas D. Huntington
- Subjects
biology ,CDKN2A ,biology.protein ,STAT5 ,Cell biology ,Progenitor - Published
- 2020
- Full Text
- View/download PDF
19. Hhex Is Essential for NK Cell Persistence by Repressing Bcl2l11-Dependent Apoptosis
- Author
-
Melissa J. Davis, Mariapia A. Degli-Esposti, Christopher E. Andoniou, Wilford Goh, Stephen L. Nutt, Soroor Hediyeh-Zadeh, Jai Rautela, Jacob T. Jackson, Nicholas D. Huntington, Matthew P. McCormack, and Rebecca B. Delconte
- Subjects
Haematopoiesis ,medicine.anatomical_structure ,Innate immune system ,BCL2L11 ,Cell growth ,Cell ,Transcriptional regulation ,medicine ,Biology ,Transcription factor ,Germline ,Cell biology - Abstract
Hhex encodes a homeobox transcriptional regulator important for embryonic development and hematopoiesis. Hhex is highly expressed in NK cells and its germline deletion results in significant defects in lymphoid development, including NK cells. However, whether Hhex is intrinsically required throughout NK cell development or for NK cell function remains unknown. To investigate this, we generated mice that specifically lack Hhex in NK cells. We found Hhex to be intrinsically required for NK cell homeostasis and subsequent in vivo viral and tumor immunity. NK cell differentiation, IL-15 responsiveness and function on a cellular level were largely normal in the absence of Hhex. Unexpectedly, increased IL-15 availability failed to rescue NK lymphopenia following conditional Hhex deletion, suggesting that Hhex regulates developmental pathways extrinsic to those dependent on IL-15. Gene expression and functional genetic approaches revealed that Hhex was required for NK cell survival by repressing BIM expression, a key apoptotic mediator in NK cells. Thus this study identifies Hhex as a novel transcription factor essential for NK cell homeostasis and immunity.
- Published
- 2020
- Full Text
- View/download PDF
20. Blind Stenanography Detection Using a Computational Immune System: A Work in Progress.
- Author
-
Jacob T. Jackson, Gregg H. Gunsch, Gary B. Lamont, and Roger L. Claypoole Jr.
- Published
- 2003
21. Hhex Regulates Hematopoietic Stem Cell Self-Renewal and Stress Hematopoiesis via Repression of Cdkn2a
- Author
-
Jacob T. Jackson, Donald Metcalf, Benjamin J. Shields, Ladina Di Rago, Wei Shi, Nicos A. Nicola, and Matthew P. McCormack
- Subjects
0301 basic medicine ,Myeloid ,medicine.medical_treatment ,Hematopoietically expressed homeobox ,Hematopoietic stem cell transplantation ,Biology ,03 medical and health sciences ,Stress, Physiological ,Cell Self Renewal ,medicine ,Animals ,Progenitor cell ,Cyclin-Dependent Kinase Inhibitor p16 ,Myeloid Progenitor Cells ,Cell Proliferation ,Homeodomain Proteins ,Hematopoietic Stem Cell Transplantation ,Hematopoietic stem cell ,Cell Biology ,Hematopoietic Stem Cells ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Cancer research ,Molecular Medicine ,Stem cell ,biology.gene ,Gene Deletion ,Transcription Factors ,Developmental Biology - Abstract
The hematopoietically expressed homeobox transcription factor (Hhex) is important for the maturation of definitive hematopoietic progenitors and B-cells during development. We have recently shown that in adult hematopoiesis, Hhex is dispensable for maintenance of hematopoietic stem cells (HSCs) and myeloid lineages but essential for the commitment of common lymphoid progenitors (CLPs) to lymphoid lineages. Here, we show that during serial bone marrow transplantation, Hhex-deleted HSCs are progressively lost, revealing an intrinsic defect in HSC self-renewal. Moreover, Hhex-deleted mice show markedly impaired hematopoietic recovery following myeloablation, due to a failure of progenitor expansion. In vitro, Hhex-null blast colonies were incapable of replating, implying a specific requirement for Hhex in immature progenitors. Transcriptome analysis of Hhex-null Lin−Sca+Kit+ cells showed that Hhex deletion leads to derepression of polycomb repressive complex 2 (PRC2) and PRC1 target genes, including the Cdkn2a locus encoding the tumor suppressors p16Ink4a and p19Arf. Indeed, loss of Cdkn2a restored the capacity of Hhex-null blast colonies to generate myeloid progenitors in vitro, as well as hematopoietic reconstitution following myeloablation in vivo. Thus, HSCs require Hhex to promote PRC2-mediated Cdkn2a repression to enable continued self-renewal and response to hematopoietic stress.
- Published
- 2017
- Full Text
- View/download PDF
22. A new lymphoid-primed progenitor marked by Dach1 downregulation identified with single cell multi-omics
- Author
-
Daniela, Amann-Zalcenstein, Luyi, Tian, Jaring, Schreuder, Sara, Tomei, Dawn S, Lin, Kirsten A, Fairfax, Jessica E, Bolden, Mark D, McKenzie, Andrew, Jarratt, Adrienne, Hilton, Jacob T, Jackson, Ladina, Di Rago, Matthew P, McCormack, Carolyn A, de Graaf, Olivia, Stonehouse, Samir, Taoudi, Warren S, Alexander, Stephen L, Nutt, Matthew E, Ritchie, Ashley P, Ng, and Shalin H, Naik
- Subjects
Mice, Knockout ,Proteomics ,Gene Expression Profiling ,Computational Biology ,High-Throughput Nucleotide Sequencing ,Mice, Transgenic ,Genomics ,Lymphoid Progenitor Cells ,Hematopoiesis ,Mice ,Animals ,Single-Cell Analysis ,Eye Proteins ,Biomarkers ,Cells, Cultured - Abstract
A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin
- Published
- 2019
23. Hhex regulates murine lymphoid progenitor survival independently of Stat5 and Cdkn2a
- Author
-
David M Tarlinton, Wilford Goh, Matthew P. McCormack, Kristy O'Donnell, Jacob T. Jackson, Nicholas D. Huntington, and Amanda Light
- Subjects
0301 basic medicine ,Cell Survival ,Immunology ,Apoptosis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,STAT5 Transcription Factor ,Immunology and Allergy ,Animals ,Lymphopoiesis ,Transcription factor ,STAT5 ,Cyclin-Dependent Kinase Inhibitor p16 ,Progenitor ,Homeodomain Proteins ,Mice, Knockout ,biology ,Hematopoietic stem cell ,Myeloid leukemia ,Lymphoid Progenitor Cells ,Cell biology ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,biology.protein ,Stem cell ,030215 immunology ,Signal Transduction ,Transcription Factors - Abstract
The transcription factor Hhex (hematopoietically expressed homeobox gene) is critical for development of multiple lymphoid lineages beyond the common lymphoid progenitor. In addition, Hhex regulates hematopoietic stem cell (HSC) self-renewal, emergency hematopoiesis, and acute myeloid leukemia initiation and maintenance. Hhex mediates its effects on HSCs and acute myeloid leukemia stem cells via repression of the Cdkn2a tumor suppressor locus. However, we report here that loss of Cdkn2a does not rescue the failure of lymphoid development caused by loss of Hhex. As loss of Hhex causes apoptosis of lymphoid progenitors associated with impaired Bcl2 expression and defective Stat5b signaling, we tested the effects of rescuing these pathways using transgenic mice. Expression of the anti-apoptotic factor Bcl2, but not activated Stat5, rescued the development of T-, B-, and NK-cell lineages in the absence of Hhex. These results indicate that Bcl2 expression, but not Stat5b signaling or loss of Cdkn2a, can overcome the lymphoid deficiencies caused by the absence of Hhex, suggesting that the primary role of this transcription factor is to promote survival of lymphoid progenitors during early lymphoid development.
- Published
- 2019
24. Hhex induces promyelocyte self-renewal and cooperates with growth factor independence to cause myeloid leukemia in mice
- Author
-
Sue Haupt, Jacob T. Jackson, Ashley P. Ng, Matthew P. McCormack, Benjamin J. Shields, and Ygal Haupt
- Subjects
0301 basic medicine ,Myeloid ,genetic structures ,Cellular differentiation ,Hematopoietically expressed homeobox ,Cell Culture Techniques ,03 medical and health sciences ,Promyelocytic leukemia protein ,Mice ,hemic and lymphatic diseases ,medicine ,Animals ,Granulocyte Precursor Cells ,Cell Self Renewal ,Cyclin-Dependent Kinase Inhibitor p16 ,Cell Nucleus ,Homeodomain Proteins ,Myeloid Neoplasia ,biology ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,Cell Differentiation ,Hematology ,medicine.disease ,Cell biology ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Leukemia, Myeloid ,Epigenetic Repression ,biology.protein ,Intercellular Signaling Peptides and Proteins ,biology.gene ,Promyelocyte ,Transcription Factors - Abstract
The hematopoietically expressed homeobox (Hhex) transcription factor is overexpressed in human myeloid leukemias. Conditional knockout models of murine acute myeloid leukemia indicate that Hhex maintains leukemia stem cell self-renewal by enabling Polycomb-mediated epigenetic repression of the Cdkn2a tumor suppressor locus, encoding p16Ink4a and p19Arf However, whether Hhex overexpression also affects hematopoietic differentiation is unknown. To study this, we retrovirally overexpressed Hhex in hematopoietic progenitors. This enabled serial replating of myeloid progenitors, leading to the rapid establishment of interleukin-3 (IL-3)-dependent promyelocytic cell lines. Use of a Hhex-ERT2 fusion protein demonstrated that continuous nuclear Hhex is required for transformation, and structure function analysis demonstrated a requirement of the DNA-binding and N-terminal-repressive domains of Hhex for promyelocytic transformation. This included the N-terminal promyelocytic leukemia protein (Pml) interaction domain, although deletion of Pml failed to prevent Hhex-induced promyelocyte transformation, implying other critical partners. Furthermore, deletion of p16Ink4a or p19Arf did not promote promyelocyte transformation, indicating that repression of distinct Hhex target genes is required for this process. Indeed, transcriptome analysis showed that Hhex overexpression resulted in repression of several myeloid developmental genes. To test the potential for Hhex overexpression to contribute to leukemic transformation, Hhex-transformed promyelocyte lines were rendered growth factor-independent using a constitutively active IL-3 receptor common β subunit (βcV449E). The resultant cell lines resulted in a rapid promyelocytic leukemia in vivo. Thus, Hhex overexpression can contribute to myeloid leukemia via multiple mechanisms including differentiation blockade and enabling epigenetic repression of the Cdkn2a locus.
- Published
- 2018
25. 2037 – DACH1 DOWNREGULATION MARKS A ‘LYMPHOID-PRIMED PROGENITOR’ IN EARLY HAEMATOPOIESIS
- Author
-
Mark D. McKenzie, Jaring Schreuder, Kirsten A. Fairfax, Andrew Jarrat, Jess Bolden, Matt McCormack, Sara Tomei, Matthew E. Ritchie, Ladina Di Rago, Adrienne A. Hilton, Daniela Zalcenstein, Jacob T. Jackson, Ashley P. Ng, Dawn S. Y. Lin, Warren S. Alexander, Shalin H. Naik, Luyi Tian, and D J Hilton
- Subjects
Cancer Research ,Myeloid ,Cell ,Cell Biology ,Hematology ,Biology ,Cell biology ,Blood cell ,Haematopoiesis ,medicine.anatomical_structure ,Downregulation and upregulation ,Megakaryocyte ,Genetics ,medicine ,Progenitor cell ,Molecular Biology ,Progenitor - Abstract
A classical view of blood cell development is that multipotent haematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. The Lin–c-kit+Sca1+Flt3+ stage, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Through single cell RNA-sequencing, we identify heterogeneous expression of Dach1 and associated genes in this fraction where it co-expressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1– subpopulation within LMPPs with lymphoid potential but devoid of myeloid potential - both in vivo and in vitro. Dach1 based separation is distinct from to separation by Rag1-GFP, Flt3, CD27, CD48 or VCAM. We term these ‘lymphoid-primed progenitors’, or LPPs. These findings define the earliest branch point of lymphoid development in haematopoiesis and a means for their prospective isolation.
- Published
- 2019
- Full Text
- View/download PDF
26. DACH1 DOWNREGULATION DELINEATE A LYMPHOID PRIMED PROGENITOR (LPP) POPULATION WITHIN THE MPP4 CLUSTER
- Author
-
Sara Tomei, Mark D. McKenzie, Shalin H. Naik, Matt McCormack, Jacob T. Jackson, D J Hilton, Ashley P. Ng, Adrienne A. Hilton, Andrew Jarratt, Ladina Di Rago, Luyi Tian, Matthew E. Ritchie, Daniela Zalcenstein, Dawn S. Lin, Ben Shields, and Jaring Schreuder
- Subjects
Cancer Research ,education.field_of_study ,Myeloid ,Population ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biology ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Megakaryocyte ,Genetics ,medicine ,Stem cell ,Progenitor cell ,education ,Molecular Biology ,Progenitor - Abstract
Classical models of haematopoiesis dictate that a single hematopoietic stem cell (HSC) with self-renewal capacity can give rise to all the hematopoietic lineage through subsequent division and loss of potential. This loss of potential is captured through surface markers that classify different stem and progenitor stages including long-term (LT-) and short-term haematopoietic stem cells (ST-HSC) that give rise to 4 categories of multipotent progenitor (MPP) subpopulations 1-4. The MPP4 cluster Lin–c-kit+Sca1+Flt3+ is also known as the lymphoid multipotent primed progenitor (LMPPs) and lack of megakaryocyte and erythroid potential. We demonstrate through cellular barcoding and scRNA-seq that the MPP4 cluster is heterogenous for fate. In particular, Dach1 expression is heterogenous and co-expressed with myeloid and stem-like genes but inversely correlates with lymphoid genes. Through the generation of a Dach1-GFP reporter mouse we demonstrate that Dach1– MPP4s represents a novel progenitor subpopulation that definitively lacks myeloid potential but retains lymphoid potential, thus termed lymphoid-primed progenitors or LPPs. This work describes the earliest definitive branch point of lymphoid development in within haematopoiesis and shows the power of scRNA-seq in identifying progenitor subpopulations.
- Published
- 2019
- Full Text
- View/download PDF
27. Requirement for Lyl1 in a model of Lmo2-driven early T-cell precursor ALL
- Author
-
Terence H. Rabbitts, Jacob T. Jackson, Matthew P. McCormack, Nicholas J. Slater, Wei Shi, David J. Curtis, Cedric S. Tremblay, Chayanica Nasa, and Benjamin J. Shields
- Subjects
LMO2 ,T-Lymphocytes ,Cellular differentiation ,Immunology ,Mice, Transgenic ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Mice ,Cancer stem cell ,Cell Line, Tumor ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Basic Helix-Loop-Helix Transcription Factors ,medicine ,Animals ,Cluster Analysis ,Humans ,T-Cell Acute Lymphocytic Leukemia Protein 1 ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,Thymocytes ,Gene Expression Regulation, Leukemic ,Gene Expression Profiling ,Cell Differentiation ,Cell Biology ,Hematology ,LIM Domain Proteins ,Gene signature ,medicine.disease ,Neoplasm Proteins ,Haematopoiesis ,Leukemia ,Cell Transformation, Neoplastic ,Neoplastic Stem Cells ,Cancer research ,TAL1 - Abstract
Lmo2 is an oncogenic transcription factor that is frequently overexpressed in T-cell acute lymphoblastic leukemia (T-ALL), including early T-cell precursor ALL (ETP-ALL) cases with poor prognosis. Lmo2 must be recruited to DNA by binding to the hematopoietic basic helix-loop-helix factors Scl/Tal1 or Lyl1. However, it is unknown which of these factors can mediate the leukemic activity of Lmo2. To address this, we have generated Lmo2-transgenic mice lacking either Scl or Lyl1 in the thymus. We show that although Scl is dispensable for Lmo2-driven leukemia, Lyl1 is critical for all oncogenic functions of Lmo2, including upregulation of a stem cell-like gene signature, aberrant self-renewal of thymocytes, and subsequent generation of T-cell leukemia. Lyl1 expression is restricted to preleukemic and leukemic stem cell populations in this model, providing a molecular explanation for the stage-specific expression of the Lmo2-induced gene expression program. Moreover, LMO2 and LYL1 are coexpressed in ETP-ALL patient samples, and LYL1 is required for growth of ETP-ALL cell lines. Thus, the LMO2-LYL1 interaction is a promising therapeutic target for inhibiting self-renewing cancer stem cells in T-ALL, including poor-prognosis ETP-ALL cases.
- Published
- 2013
- Full Text
- View/download PDF
28. CD8α+ DCs can be induced in the absence of transcription factors Id2, Nfil3, and Batf3
- Author
-
Gabrielle T. Belz, Jacob T. Jackson, Cyril Seillet, Kelli P. A. MacDonald, Stephen L. Nutt, Kate A. Markey, Hugh J.M. Brady, and Geoffrey R. Hill
- Subjects
XCR1 ,Cell Survival ,CD8 Antigens ,Cellular differentiation ,Immunology ,Antigen presentation ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Herpesvirus 1, Human ,Biology ,Biochemistry ,Mice ,Cross-Priming ,Animals ,Antigens, Viral ,Transcription factor ,Cells, Cultured ,Inhibitor of Differentiation Protein 2 ,NFIL3 ,Cross-presentation ,Cell Differentiation ,hemic and immune systems ,Dendritic Cells ,Cell Biology ,Hematology ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,Basic-Leucine Zipper Transcription Factors ,Interferon Regulatory Factors ,IRF8 ,Transcription Factors ,Interferon regulatory factors - Abstract
Antiviral immunity and cross-presentation is mediated constitutively through CD8α+ and CD103+ DCs. Development of these DC subsets is thought to require the transcription factors Irf8, Id2, Nfil3, and Batf3, although how this network is regulated is poorly defined. We addressed the nature of the differentiation blocks observed in the absence of these factors and found that although all 4 factors are required for CD103+ DC development, only Irf8 is essential for CD8α+ DCs. CD8α+ DCs emerged in the absence of Id2, Nfil3 and Batf3 in short-term bone marrow reconstitution. These “induced” CD8α+ DCs exhibit several hallmarks of classic CD8α+ DCs including the expression of CD24, Tlr3, Xcr1, Clec9A, and the capacity to cross-present soluble, cell-associated antigens and viral antigens even in the absence of Batf3. Collectively, these results uncover a previously undescribed pathway by which CD8α+ DCs emerge independent of Id2, Nfil3, and Batf3, but dependent on Irf8.
- Published
- 2013
- Full Text
- View/download PDF
29. Rediscovery of the Headwater Catfish Ictalurus lupus (Ictaluridae) in a Western Gulf-Slope Drainage
- Author
-
Jacob T. Jackson, Timothy H. Bonner, Diana J. McHenry, Michael R. J. Forstner, and Preston T. Bean
- Subjects
education.field_of_study ,geography ,geography.geographical_feature_category ,biology ,Headwater catfish ,Range (biology) ,Ecology ,Population ,biology.organism_classification ,Fishery ,Ictalurus ,Spring (hydrology) ,education ,Ecology, Evolution, Behavior and Systematics ,Ictaluridae ,Channel (geography) ,Catfish - Abstract
Stream flow modifications, such as reduced spring flow and construction of low-head dams, have led to declines in the headwater catfish (Ictalurus lupus) across much of its range, as has competition with channel catfish (I. punctatus). Hybridization between headwater catfish and channel catfish also poses a threat to the headwater catfish. Our analyses of cytochrome-b sequences of headwater catfish from the Frio River, Devils River, and Independence Creek indicated that a population of headwater catfish occurs in the Frio River of the Nueces Drainage, where it was considered extirpated since 1967, and that hybridization is occurring in populations that inhabit the Frio River and Independence Creek. No obvious sign of hybridization was present in the population of headwater catfish in the Devils River.
- Published
- 2011
- Full Text
- View/download PDF
30. Id2 expression delineates differential checkpoints in the genetic program of CD8α+and CD103+dendritic cell lineages
- Author
-
Ruijie Liu, Li Wu, Axel Kallies, Gabrielle T. Belz, Annie Xin, Mary J Camilleri, Gordon K. Smyth, Frederick Masson, Yifang Hu, Stephen L. Nutt, Jacob T. Jackson, Angela D'Amico, Sebastian Carotta, and Adele M. Mount
- Subjects
0303 health sciences ,General Immunology and Microbiology ,General Neuroscience ,Cellular differentiation ,Antigen presentation ,Priming (immunology) ,Cross-presentation ,hemic and immune systems ,chemical and pharmacologic phenomena ,Dendritic cell ,Biology ,Acquired immune system ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunology ,Molecular Biology ,Transcription factor ,030304 developmental biology ,030215 immunology - Abstract
Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α+ and CD103+ DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103+ and CD8α+ lineages. Notably, CD103+ DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α− DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP+ DCs. It did not restore cross-presenting capacity to Batf3−/− or CD103−Sirp-α−DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.
- Published
- 2011
- Full Text
- View/download PDF
31. The NUP98-HOXD13 Fusion Oncogene Induces Thymocyte Self-Renewal Via Lmo2/Lyl1
- Author
-
David J. Curtis, Anh Vo, Benjamin J. Shields, Wei Shi, Matthew P. McCormack, Jacob T. Jackson, and Christopher Slape
- Subjects
LMO2 ,Fusion ,Oncogene ,Chemistry ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Cell biology ,Transplantation ,Thymocyte ,Haematopoiesis ,LYL1 ,hemic and lymphatic diseases ,Transcription factor - Abstract
T-cell Acute Lymphoblastic Leukaemias (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. Whilst it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukemia (AML) as well as T-ALL. We find that thymocytes from preleukemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional program closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression program in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus, multiple T-cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1, however NHD13 can also promote T-ALL via an alternative pathway. Disclosures Curtis: MERCK: Membership on an entity's Board of Directors or advisory committees; CRC Cancer Therapeutics: Patents & Royalties, Research Funding.
- Published
- 2018
- Full Text
- View/download PDF
32. Hhex Promotes Lymphoid Progenitor Survival Independently of Stat5 and Cdkn2a
- Author
-
Amanda Light, David M. Tarlinton, Matthew P. McCormack, Kristy O'Donnell, Nicholas D. Huntington, Wil Goh, and Jacob T. Jackson
- Subjects
Cancer Research ,biology ,CDKN2A ,Genetics ,Cancer research ,biology.protein ,Cell Biology ,Hematology ,Molecular Biology ,STAT5 ,Progenitor - Published
- 2018
- Full Text
- View/download PDF
33. A Mitochondrial DNA Phylogeny of Extant Species of the GenusTrachemyswith Resulting Taxonomic Implications
- Author
-
Robert W. Guthrie, Michael R. J. Forstner, Jacob T. Jackson, and David E. Starkey
- Subjects
Mitochondrial DNA ,Taxon ,biology ,Phylogenetic tree ,Phylogenetics ,Zoology ,Animal Science and Zoology ,Trachemys ,Taxonomy (biology) ,Subspecies ,biology.organism_classification ,Ecology, Evolution, Behavior and Systematics ,Maximum parsimony - Abstract
The phylogenetic relationships among taxa within the emydid genus Trachemys have largely remained unresolved. A 768-basepair fragment of ND4, as well as the histidine, serine, and leucine tRNAs were sequenced from 18 of 26 of the extant species and subspecies of Trachemys. The aligned sequences were analyzed using maximum parsimony, maximum likelihood, and Bayesian methods. The results support the taxonomy of the genus as proposed by Seidel.
- Published
- 2008
- Full Text
- View/download PDF
34. Detection of Salmonellae in Captive and Free-Ranging Turtles Using Enrichment Culture and Polymerase Chain Reaction
- Author
-
Francis L. Rose, Dittmar Hahn, James P. Gaertner, Jacob T. Jackson, and Michael R. J. Forstner
- Subjects
Claw ,Salmonella ,Trachemys gaigeae ,biology ,Free ranging ,Ecology ,Zoology ,biology.organism_classification ,medicine.disease_cause ,Enrichment culture ,law.invention ,law ,medicine ,Apalone spinifera ,Animal Science and Zoology ,Carapace ,Ecology, Evolution, Behavior and Systematics ,Polymerase chain reaction - Abstract
Information on the importance of captive turtles as sources of human Salmonella infections is well established; however, data on the potential of free-ranging turtles as carriers of salmonellae are scarce and contradictory. We combined traditional culture techniques and molecular tools to analyze swabs obtained from the cloacae and from different body parts of captive and free-ranging turtles for salmonellae. Salmonellae were detected in 50% of captive turtles (N = 10). A similar percentage of detection (51%) was obtained for salmonellae in free-ranging turtles from the Rio Grande (N = 80) with six sites at Big Bend National Park, Texas, and one site at Elephant Butte Reservoir, New Mexico, analyzed. Here, 46% of Trachemys gaigeae (N = 36), 56% of Apalone spinifera (N = 43), and the only individual of Chrysemys picta were positive for salmonellae. These percentages of detection of salmonellae in turtles were independent of the location of the sampling in the Rio Grande, the species and the gender of the turtles. Although individuals of captive turtles testing positive for salmonellae were generally positive for all body parts tested (i.e., the cloacae, the carapace, the ventral base of the left rear leg, underneath one or more of the claws on the front feet, and the ventral base of the tail), individuals of free-ranging turtles testing positive for salmonellae were often positive for only one or two body parts. Our results demonstrate that salmonellae are prevalent in high rates in both captive and free-ranging turtles.
- Published
- 2008
- Full Text
- View/download PDF
35. Adoptive Transfer of Chimeric Fc ε RI Receptor Gene-Modified Human T Cells for Cancer Immunotherapy
- Author
-
Jacob T. Jackson, Michele W.L. Teng, Mark J. Smyth, Michael H. Kershaw, and Phillip K. Darcy
- Subjects
Adoptive cell transfer ,biology ,T cell ,Fc receptor ,CD28 ,Immunoglobulin E ,Molecular biology ,medicine.anatomical_structure ,Antigen ,Genetics ,biology.protein ,medicine ,Molecular Medicine ,Cytotoxic T cell ,Antibody ,Molecular Biology - Abstract
Immunotherapeutic approaches involving genetic modification of T cells show promise in generating highly specific tumor-reactive effector cells for cancer treatment. Given the high affinity of Fc e RI (the subtype I Fc receptor for IgE) for IgE monoclonal antibody (mAb), modification of T cells with chimeric Fc e RI in combination with tumor-specific IgE mAbs is potentially a powerful and effective strategy to specifically target T cells to tumor cells. In this study, we retrovirally transduce human primary T cells with a cDNA encoding the extracellular domain of Fc e RI linked to the hinge and transmembrane domains of Fc γ RI and the cytoplasmic domains of CD28 and T cell receptor ζ chain (Fc e RI-CD28-ζ). We demonstrate that human T cells expressing Fc e RI-CD28-ζ, in the presence of tumor-specific IgE mAb recognizing mouse CD8 antigen (Ly- 2.1+), can specifically secrete cytokine, proliferate, and mediate cytotoxic function after antigen ligation. Furthermore, adoptive transfer of Fc e RI-CD28-ζ cells ...
- Published
- 2006
- Full Text
- View/download PDF
36. Adoptive transfer of gene-engineered CD4+ helper T cells induces potent primary and secondary tumor rejection
- Author
-
Joseph A. Trapani, Michele W.L. Teng, Nicole M. Haynes, Loretta Cerutti, Shayna E. A. Street, Maria Moeller, Jacob T. Jackson, Michael H. Kershaw, Stephen M. Jane, Phillip K. Darcy, and Mark J. Smyth
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,Recombinant Fusion Proteins ,Immunology ,Receptors, Cell Surface ,CD8-Positive T-Lymphocytes ,Biology ,Biochemistry ,Interferon-gamma ,Mice ,Interleukin 21 ,Antigen ,Cell Line, Tumor ,Neoplasms ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Cell Proliferation ,Mice, Knockout ,Mice, Inbred BALB C ,ZAP70 ,T-Lymphocytes, Helper-Inducer ,Cell Biology ,Hematology ,Adoptive Transfer ,Survival Rate ,Gene Expression Regulation ,Cancer research ,Cytokines ,Genetic Engineering ,CD8 ,Single-Chain Antibodies - Abstract
Because CD4+ T cells play a key role in aiding cellular immune responses, we wanted to assess whether increasing numbers of gene-engineered antigen-restricted CD4+ T cells could enhance an antitumor response mediated by similarly gene-engineered CD8+ T cells. In this study, we have used retroviral transduction to generate erbB2-reactive mouse T-cell populations composed of various proportions of CD4+ and CD8+ cells and then determined the antitumor reactivity of these mixtures. Gene-modified CD4+ and CD8+ T cells were shown to specifically secrete Tc1 (T cytotoxic-1) or Tc2 cytokines, proliferate, and lyse erbB2+ tumor targets following antigen ligation in vitro. In adoptive transfer experiments using severe combined immunodeficient (scid) mice, we demonstrated that injection of equivalent numbers of antigenspecific engineered CD8+ and CD4+ T cells led to significant improvement in survival of mice bearing established lung metastases compared with transfer of unfractionated (largely CD8+) engineered T cells. Transferred CD4+ T cells had to be antigen-specific (not just activated) and secrete interferon γ (IFN-γ) to potentiate the antitumor effect. Importantly, antitumor responses in these mice correlated with localization and persistence of geneengineered T cells at the tumor site. Strikingly, mice that survived primary tumor challenge could reject a subsequent rechallenge. Overall, this study has highlighted the therapeutic potential of using combined transfer of antigen-specific gene-modified CD8+ and CD4+ T cells to significantly enhance T-cell adoptive transfer strategies for cancer therapy.
- Published
- 2005
- Full Text
- View/download PDF
37. The Functional Basis for Hemophagocytic Lymphohistiocytosis in a Patient with Co-inherited Missense Mutations in the Perforin (PFN1) Gene
- Author
-
Joseph A. Trapani, Ilia Voskoboinik, Jacob T. Jackson, Marie Claude Thia, Mark J. Smyth, Erika Cretney, Phillip K. Darcy, Stephen M. Jane, Kylie A. Browne, and Annette De Bono
- Subjects
Pore Forming Cytotoxic Proteins ,cytotoxic granule ,Histiocytosis, Non-Langerhans-Cell ,Immunology ,Mutation, Missense ,chemical and pharmacologic phenomena ,Biology ,Basophil ,medicine.disease_cause ,Jurkat cells ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,NK cell ,Hemophagocytic lymphohistiocytosis ,Mutation ,Membrane Glycoproteins ,Perforin ,Brief Definitive Report ,hemic and immune systems ,medicine.disease ,Molecular biology ,Rats ,Granzyme B ,Leukemia ,medicine.anatomical_structure ,CTL ,biology.protein ,immunodeficiency ,T-Lymphocytes, Cytotoxic ,HLH - Abstract
About 30% of cases of the autosomal recessive immunodeficiency disorder hemophagocytic lymphohistiocytosis are believed to be caused by inactivating mutations of the perforin gene. We expressed perforin in rat basophil leukemia cells to define the basis of perforin dysfunction associated with two mutations, R225W and G429E, inherited by a compound heterozygote patient. Whereas RBL cells expressing wild-type perforin (67 kD) efficiently killed Jurkat target cells to which they were conjugated, the substitution to tryptophan at position 225 resulted in expression of a truncated (∼45 kD) form of the protein, complete loss of cytotoxicity, and failure to traffic to rat basophil leukemia secretory granules. By contrast, G429E perforin was correctly processed, stored, and released, but the rat basophil leukemia cells possessed reduced cytotoxicity. The defective function of G429E perforin mapped downstream of exocytosis and was due to its reduced ability to bind lipid membranes in a calcium-dependent manner. This study elucidates the cellular basis for perforin dysfunctions in hemophagocytic lymphohistiocytosis and provides the means for studying structure–function relationships for lymphocyte perforin.
- Published
- 2004
- Full Text
- View/download PDF
38. A functional role for CD28 costimulation in tumor recognition by single-chain receptor-modified T cells
- Author
-
Maria Moeller, Loretta Cerutti, Michael H. Kershaw, Jacob T. Jackson, Michele W.L. Teng, Stephen M. Jane, Jane E. Tanner, Nicole M. Haynes, Joseph A. Trapani, Phillip K. Darcy, and Mark J. Smyth
- Subjects
Interleukin 2 ,Cancer Research ,Receptor, ErbB-2 ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Immunoglobulin Variable Region ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,Mice ,CD28 Antigens ,Antigen ,Transduction, Genetic ,Cell Line, Tumor ,medicine ,Animals ,Humans ,IL-2 receptor ,Receptor ,Molecular Biology ,Mice, Inbred BALB C ,CD28 ,hemic and immune systems ,Neoplasms, Experimental ,respiratory system ,Molecular biology ,Chimeric antigen receptor ,Cell biology ,Retroviridae ,Cytokine ,medicine.anatomical_structure ,Amino Acid Substitution ,Molecular Medicine ,medicine.drug - Abstract
T cells engineered to express single-chain antibody receptors that incorporate TCR-zeta and cluster designation (CD)28 signaling domains (scFv-alpha-erbB2-CD28-zeta) can be redirected in vivo to cancer cells that lack triggering costimulatory molecules. To assess the contribution of CD28 signaling to the function of the scFv-CD28-zeta receptor, we expressed a series of mutated scFv-CD28-zeta receptors directed against erbB2. Residues known to be critical for CD28 signaling were mutated from tyrosine to phenylalanine at position 170 or proline to alanine at positions 187 and 190. Primary mouse T cells expressing either of the mutant receptors demonstrated impaired cytokine (IFN-gamma and GM-CSF) production and decreased proliferation after antigen ligation in vitro and decreased antitumor efficacy in vivo compared with T cells expressing the wild-type scFv-CD28-zeta receptor, suggesting a key signaling role for the CD28 component of the scFv-CD28-zeta receptor. Importantly, cell surface expression, binding capacity and cytolytic activity mediated by the scFv-CD28-zeta receptor were not diminished by either mutation. Overall, this study has definitively demonstrated a functional role for the CD28 component of the scFv-CD28-zeta receptor and has shown that incorporation of costimulatory activity in chimeric scFv receptors is a powerful approach for improving adoptive cancer immunotherapy.
- Published
- 2004
- Full Text
- View/download PDF
39. Rejection of Syngeneic Colon Carcinoma by CTLs Expressing Single-Chain Antibody Receptors Codelivering CD28 Costimulation
- Author
-
Nicole M. Haynes, Michael H. Kershaw, Joseph A. Trapani, Stephen M. Jane, Mark J. Smyth, Loretta Cerruti, Phillip K. Darcy, Jacob T. Jackson, and Michele W.L. Teng
- Subjects
Cytotoxicity, Immunologic ,Graft Rejection ,Pore Forming Cytotoxic Proteins ,Recombinant Fusion Proteins ,T cell ,Immunology ,Immunoglobulin Variable Region ,Receptors, Antigen, T-Cell ,Epitopes, T-Lymphocyte ,chemical and pharmacologic phenomena ,Mice, SCID ,Adenocarcinoma ,Biology ,Major histocompatibility complex ,Immunotherapy, Adoptive ,Epitope ,Interferon-gamma ,Mice ,Adjuvants, Immunologic ,CD28 Antigens ,Antigen ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Receptors, Immunologic ,Receptor ,Cells, Cultured ,Mice, Knockout ,Mice, Inbred BALB C ,Membrane Glycoproteins ,Perforin ,Membrane Proteins ,CD28 ,T lymphocyte ,Molecular biology ,Growth Inhibitors ,Carcinoembryonic Antigen ,Mice, Inbred C57BL ,Transplantation, Isogeneic ,medicine.anatomical_structure ,Colonic Neoplasms ,biology.protein ,Cancer research ,Binding Sites, Antibody ,T-Lymphocytes, Cytotoxic - Abstract
A new strategy to improve the therapeutic utility of redirected T cells for cancer involves the development of novel Ag-specific chimeric receptors capable of stimulating optimal and sustained T cell antitumor activity in vivo. Given that T cells require both primary and costimulatory signals for optimal activation and that many tumors do not express critical costimulatory ligands, modified single-chain Ab receptors have been engineered to codeliver CD28 costimulation. In this study, we have compared the antitumor potency of primary T lymphocytes expressing carcinoembryonic Ag (CEA)-reactive chimeric receptors that incorporate either TCR-ζ or CD28/TCR-ζ signaling. Although both receptor-transduced T cell effector populations demonstrated cytolysis of CEA+ tumors in vitro, T cells expressing the single-chain variable fragment of Ig (scFv)-CD28-ζ chimera had a far greater capacity to control the growth of CEA+ xenogeneic and syngeneic colon carcinomas in vivo. The observed enhanced antitumor activity of T cells expressing the scFv-CD28-ζ receptor was critically dependent on perforin and the production of IFN-γ. Overall, this study has illustrated the ability of a chimeric scFv receptor capable of harnessing the signaling machinery of both TCR-ζ and CD28 to augment T cell immunity against tumors that have lost expression of both MHC/peptide and costimulatory ligands in vivo.
- Published
- 2002
- Full Text
- View/download PDF
40. Inferring Absence of Houston Toads Given Imperfect Detection Probabilities
- Author
-
Jacob T. Jackson, Floyd W. Weckerly, Todd M. Swannack, and Michael R. J. Forstner
- Subjects
Ecology ,Wildlife ,Endangered species ,Effective management ,Fishery ,Geography ,Single species ,Geological survey ,Seasonal breeder ,General Earth and Planetary Sciences ,%22">Fish ,Bufo houstonensis ,Ecology, Evolution, Behavior and Systematics ,Nature and Landscape Conservation ,General Environmental Science - Abstract
Researchers commonly use auditory surveys to determine the presence of anuran species (Bridges and Dorcas 2000, Crouch and Paton 2002, Schmidt 2003, Pierce and Gutzweiller 2004, U.S. Geological Survey 2005). Researchers conduct the surveys during the breeding season when the species of interest are vocalizing to attract mates. Researchers record environmental conditions and listen for the call of interest. In one such method, trained observers use auditory cues alone to detect and monitor the presence or absence of endangered Houston toads (Bufo houstonensis; Forstner and Swannack 2004). The Houston toad is endemic to eastcentral Texas, USA, and was first listed as an endangered species in 1970. Observers in this program train for several seasons in detection and differentiation of calls, and they must have certification from the United States Fish and Wildlife Service (USFWS). However, one problem with any in situ sampling method is that detection of organisms is imperfect (Kery 2002, MacKenzie et al. 2002, Schmidt 2003, Bailey et al. 2004). A conclusion of absence may simply be an artifact of the survey scheme, rather than actual absence. The USFWS provides explicit guidelines for audio call surveys to federally permitted individuals. The guidelines delineate the appropriate season, weather conditions, duration, and number of surveys required in a season. One of the specified guidelines is the completion of, at minimum, 6 survey nights during suitable season and weather conditions. The ability to detect Houston toads reliably with this survey protocol is a concern of the USFWS and necessary for effective management of this species. The number of auditory surveys needed to determine the presence or absence of a single species, the Houston toad, accurately is the focus of this study. This is a pressing concern when monitoring an endangered species such as the Houston toad. We estimated the probability of detecting Houston toads when present (detection probability) and used these probabilities to determine the number of surveys required to be confident in a verdict of Houston toad absence from potential breeding sites. Study Area
- Published
- 2006
- Full Text
- View/download PDF
41. Id2 expression delineates differential checkpoints in the genetic program of CD8α+ and CD103+ dendritic cell lineages
- Author
-
Jacob T, Jackson, Yifang, Hu, Ruijie, Liu, Frederick, Masson, Angela, D'Amico, Sebastian, Carotta, Annie, Xin, Mary J, Camilleri, Adele M, Mount, Axel, Kallies, Li, Wu, Gordon K, Smyth, Stephen L, Nutt, and Gabrielle T, Belz
- Subjects
Mice, Inbred BALB C ,CD8 Antigens ,Gene Expression ,Cell Differentiation ,Mice, Transgenic ,Dendritic Cells ,Id2 ,Models, Biological ,Article ,Genes, cdc ,Mice, Inbred C57BL ,Mice ,antigen presentation ,differentiaton ,Antigens, CD ,transcription factors ,Animals ,Cell Lineage ,priming ,Integrin alpha Chains ,Cells, Cultured ,cross-presentation ,Inhibitor of Differentiation Protein 2 - Abstract
Id2 expression delineates differential checkpoints in the genetic program of CD8α+ and CD103+ dendritic cell lineages A novel reporter mouse for the transcription factor inhibitor of DNA binding-2 (Id2) provides insight into the differentiation of DC subsets in vivo and in vitro., Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α+ and CD103+ DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103+ and CD8α+ lineages. Notably, CD103+ DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α− DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP+ DCs. It did not restore cross-presenting capacity to Batf3−/− or CD103−Sirp-α−DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.
- Published
- 2011
42. Adoptive transfer of gene-modified primary NK cells can specifically inhibit tumor progression in vivo
- Author
-
Phillip K. Darcy, Hollie J Pegram, Michael H. Kershaw, Jacob T. Jackson, and Mark J. Smyth
- Subjects
Cytotoxicity, Immunologic ,Adoptive cell transfer ,Lymphoma ,Receptor, ErbB-2 ,T cell ,medicine.medical_treatment ,Recombinant Fusion Proteins ,Immunology ,Immunoglobulin Variable Region ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Immunotherapy, Adoptive ,Antibodies ,Interleukin 21 ,Mice ,Cancer immunotherapy ,CD28 Antigens ,MHC class I ,medicine ,Immunology and Allergy ,Animals ,Humans ,Lymphokine-activated killer cell ,biology ,Janus kinase 3 ,Gene Transfer Techniques ,Molecular biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,Survival Rate ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Interleukin 12 ,Disease Progression - Abstract
NK cells hold great potential for improving the immunotherapy of cancer. Nevertheless, tumor cells can effectively escape NK cell-mediated apoptosis through interaction of MHC molecules with NK cell inhibitory receptors. Thus, to harness NK cell effector function against tumors, we used Amaxa gene transfer technology to gene-modify primary mouse NK cells with a chimeric single-chain variable fragment (scFv) receptor specific for the human erbB2 tumor-associated Ag. The chimeric receptor was composed of the extracellular scFv anti-erbB2 Ab linked to the transmembrane and cytoplasmic CD28 and TCR-ζ signaling domains (scFv-CD28-ζ). In this study we demonstrated that mouse NK cells gene-modified with this chimera could specifically mediate enhanced killing of an erbB2+ MHC class I+ lymphoma in a perforin-dependent manner. Expression of the chimera did not interfere with NK cell-mediated cytotoxicity mediated by endogenous NK receptors. Furthermore, adoptive transfer of gene-modified NK cells significantly enhanced the survival of RAG mice bearing established i.p. RMA-erbB2+ lymphoma. In summary, these data suggest that use of genetically modified NK cells could broaden the scope of cancer immunotherapy for patients.
- Published
- 2008
43. The critical role of the colony-stimulating factor-1 receptor in the differentiation of myeloblastic leukemia cells
- Author
-
Nicholas J. Wilson, Paul J. Masendycz, Glen M. Scholz, John A. Hamilton, Genevieve Whitty, Dominic De Nardo, and Jacob T. Jackson
- Subjects
Macrophage colony-stimulating factor ,Cancer Research ,Receptors, OSM-LIF ,Interleukin-6 ,Cellular differentiation ,Macrophages ,Priming (immunology) ,Cell Differentiation ,Receptor, Macrophage Colony-Stimulating Factor ,Biology ,Leukemia Inhibitory Factor ,Colony stimulating factor 1 receptor ,Haematopoiesis ,Leukemia, Myeloid, Acute ,Mice ,Oncology ,Cell Line, Tumor ,Cancer research ,Animals ,Signal transduction ,Kinase activity ,Molecular Biology ,Leukemia inhibitory factor - Abstract
How diverse stimuli control hemopoietic lineage development is unknown. An early event during induction of macrophage differentiation in the myeloblastic leukemia M1 cell line by different stimuli, such as leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), is expression of the colony-stimulating factor-1 receptor (CSF-1R). We report that expression of active CSF-1R in M1 cells accelerated their subsequent terminal differentiation into macrophages in response to LIF and IL-6 when compared with cells lacking the CSF-1R or expressing the receptor with compromised kinase activity; however, there was no requirement for signaling through the CSF-1R, for example, via endogenous CSF-1, during the actual LIF-induced and IL-6–induced differentiation stage. Differences were noted in the signaling pathways downstream of the LIF receptor depending on the presence of the CSF-1R. Both LIF and IL-6 gave an additive response with CSF-1, consistent with LIF and IL-6 acting via a different signaling pathway (signal transducer and activator of transcription 3 dependent) than CSF-1 (extracellular signal-regulated kinase dependent). Based at least on this cell model, we propose that terminal macrophage differentiation involves a critical priming or deterministic phase in which signaling by the CSF-1R prepares a precursor population for subsequent rapid terminal macrophage differentiation by diverse stimuli. We also propose that expression and activation of the CSF-1R explain much prior literature on macrophage lineage commitment in M1 leukemic cells and may be important in controlling the progression of certain myeloid leukemias. (Mol Cancer Res 2008;6(3):458–67)
- Published
- 2008
44. Adoptive transfer of chimeric FcepsilonRI gene-modified human T cells for cancer immunotherapy
- Author
-
Michele W L, Teng, Michael H, Kershaw, Jacob T, Jackson, Mark J, Smyth, and Phillip K, Darcy
- Subjects
Base Sequence ,Thymoma ,Receptors, IgE ,T-Lymphocytes ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,Membrane Proteins ,Immunoglobulin E ,Cytotoxicity Tests, Immunologic ,Adoptive Transfer ,Cancer Vaccines ,Recombinant Proteins ,Cell Line ,Survival Rate ,Disease Models, Animal ,Mice ,CD28 Antigens ,Transduction, Genetic ,Animals ,Cytokines ,Humans ,Immunotherapy ,Cell Proliferation - Abstract
Immunotherapeutic approaches involving genetic modification of T cells show promise in generating highly specific tumor-reactive effector cells for cancer treatment. Given the high affinity of FcRI (the subtype I Fc receptor for IgE) for IgE monoclonal antibody (mAb), modification of T cells with chimeric FcRI in combination with tumor-specific IgE mAbs is potentially a powerful and effective strategy to specifically target T cells to tumor cells. In this study, we retrovirally transduce human primary T cells with a cDNA encoding the extracellular domain of FcRI linked to the hinge and transmembrane domains of FcRI and the cytoplasmic domains of CD28 and T cell receptor zeta chain (FcRI-CD28-zeta). We demonstrate that human T cells expressing FcRI-CD28-zeta, in the presence of tumor-specific IgE mAb recognizing mouse CD8 antigen (Ly- 2.1+), can specifically secrete cytokine, proliferate, and mediate cytotoxic function after antigen ligation. Furthermore, adoptive transfer of FcRI-CD28-zeta cells incubated with anti-Ly-2.1 IgE mAb significantly enhances the survival of irradiated nonobese diabetic-severe combined immunodeficiency mice bearing Ly-2.1+ tumor compared with control mice. Thus, this set of experiments demonstrates that Fc gene-engineered human T cells mediate effector function in vitro and in vivo in an IgE-dependent manner and thus a novel and valid approach for cancer therapy can now be further developed.
- Published
- 2006
45. Gene-engineered T cells as a superior adjuvant therapy for metastatic cancer
- Author
-
Yoshihiro Hayakawa, Jane E. Tanner, Rachel Cameron, Jacob T. Jackson, Mark J. Smyth, Phillip K. Darcy, Shayna E. A. Street, Michele W.L. Teng, Joseph A. Trapani, Nicole M. Haynes, Maria Moeller, and Michael H. Kershaw
- Subjects
Oncology ,Lung Neoplasms ,Receptor, ErbB-2 ,medicine.medical_treatment ,T-Cell Antigen Receptor Specificity ,Mice, SCID ,Immunotherapy, Adoptive ,Mice ,T-Lymphocyte Subsets ,Immunology and Allergy ,Medicine ,skin and connective tissue diseases ,Mice, Inbred BALB C ,Antibiotics, Antineoplastic ,Liver Neoplasms ,Antibodies, Monoclonal ,Metastatic breast cancer ,Primary tumor ,Combined Modality Therapy ,Neoplasm Proteins ,medicine.anatomical_structure ,Chemotherapy, Adjuvant ,Female ,Fluorouracil ,Genetic Engineering ,Adjuvant ,medicine.medical_specialty ,Antimetabolites, Antineoplastic ,T cell ,Recombinant Fusion Proteins ,Immunology ,Receptors, Antigen, T-Cell ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Interferon-gamma ,Breast cancer ,Internal medicine ,Adjuvant therapy ,Animals ,Humans ,business.industry ,Carcinoma ,Cancer ,Mammary Neoplasms, Experimental ,Membrane Proteins ,Immunotherapy ,Trastuzumab ,medicine.disease ,Doxorubicin ,Drug Resistance, Neoplasm ,business ,Neoplasm Transplantation - Abstract
The major limiting factor in the successful application of adjuvant therapy for metastatic disease is the lack of adjuvant specificity that leads to severe side effects. Reasoning that T cells of the immune system are highly specific, we generated tumor-specific T cells by genetic modification of mouse primary T cells with a chimeric receptor reactive with the human breast cancer-associated Ag erbB-2. These T cells killed breast cancer cells and secreted IFN-γ in an Ag-specific manner in vitro. We investigated their use against metastatic breast cancer in mice in an adjuvant setting, and compared their effectiveness with the commonly applied adjuvants doxorubicin, 5-fluorouracil, and herceptin. Mice were inoculated orthotopically with the human erbB-2-expressing spontaneously metastatic mouse breast cancer 4T1.2 in mammary tissue, and the primary tumor was surgically removed 8 days later. Significant metastatic disease was demonstrated in lung and liver at the time of surgery on day 8 with increased tumor burden at later time points. T cell adjuvant treatment of day 8 metastatic disease resulted in dramatic increases in survival of mice, and this survival was significantly greater than that afforded by either doxorubicin, 5-fluorouracil, or herceptin.
- Published
- 2004
46. Mutational analysis of P-glycoprotein: suppression of caspase activation in the absence of ATP-dependent drug efflux
- Author
-
Ricky W. Johnstone, Phillip K. Darcy, Jacob T. Jackson, Jane E. Tanner, Stephen M. Jane, Mark J. Smyth, Loretta Cerruti, and Kellie M. Tainton
- Subjects
Time Factors ,Lymphoma ,Cell Survival ,DNA Mutational Analysis ,Green Fluorescent Proteins ,Mitosis ,Antineoplastic Agents ,Apoptosis ,Hydroxamic Acids ,Cell Line ,Structure-Activity Relationship ,Adenosine Triphosphate ,Cell Line, Tumor ,Humans ,ATP Binding Cassette Transporter, Subfamily B, Member 1 ,Molecular Biology ,Mitotic catastrophe ,Caspase ,P-glycoprotein ,Adenosine Triphosphatases ,Cell Nucleus ,biology ,Cytochrome c ,Hydrolysis ,Cell Membrane ,Cytochromes c ,Cell Biology ,Antineoplastic Agents, Phytogenic ,Drug Resistance, Multiple ,Cell biology ,Enzyme Activation ,Retroviridae ,Cell culture ,Drug Resistance, Neoplasm ,Vincristine ,Caspases ,Mutation ,biology.protein ,Efflux ,Idarubicin - Abstract
P-glycoprotein (P-gp) can induce multidrug resistance (MDR) through the ATP-dependent efflux of chemotherapeutic agents. We have previously shown that P-gp can inhibit nondrug apoptotic stimuli by suppressing the activation of caspases. To determine if this additional activity is functionally linked to ATP hydrolysis, we expressed wild-type and ATPase-mutant P-gp and showed that cells expressing mutant P-gp could not efflux chemotherapeutic drugs but remained relatively resistant to apoptosis. CEM lymphoma cells expressing mutant P-gp treated with vincristine showed a decrease in the fraction of cells with apoptotic morphology, cytochrome c release from the mitochondria and suppression of caspase activation, yet still accumulated in mitosis and showed a loss of clonogenic potential. The loss of clonogenicity in vincristine-treated cells expressing mutant P-gp was associated with accumulation of cells in mitosis and the presence of multinucleated cells consistent with mitotic catastrophe. The antiapoptotic effect of mutant P-gp was not affected by antibodies that inhibit the efflux function of the protein. These data are consistent with a dual activity model for P-gp-induced MDR involving both ATPase-dependent drug efflux and ATPase-independent inhibition of apoptosis. The structure-function analyses described herein provide novel insight into the mechanisms of action of P-gp in mediating MDR.
- Published
- 2004
47. Wavelet-based steganalysis using a computational immune system approach
- Author
-
Gregg H. Gunsch, Roger L. Claypoole, Gary B. Lamont, and Jacob T. Jackson
- Subjects
Steganalysis ,Wavelet ,Steganography ,Computer science ,Genetic algorithm ,ComputingMethodologies_IMAGEPROCESSINGANDCOMPUTERVISION ,Wavelet transform ,Leverage (statistics) ,Image processing ,Data mining ,computer.software_genre ,computer ,Signature (logic) - Abstract
Current signature-based approaches for detecting the presence of hidden messages in digital files - the initial step in steganalysis - rely on discerning "footprints" of steganographic tools. Of greater recent concern is detecting the use of novel proprietary or "home-grown" tools for which no signature has been established. This research focuses on detecting anomalies in seemingly innocuous images, applying a genetic algorithm within a computational immune system to leverage powerful image processing through wavelet analysis. The sensors developed with this new, synergistic system demonstrated a surprising level of capability to detect the use of steganographic tools for which the system had no previous exposure.© (2003) COPYRIGHT SPIE--The International Society for Optical Engineering. Downloading of the abstract is permitted for personal use only.
- Published
- 2003
- Full Text
- View/download PDF
48. Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors
- Author
-
Stephen M. Jane, Michael H. Kershaw, Nicole M. Haynes, Michele W.L. Teng, Phillip K. Darcy, Mark J. Smyth, Loretta Cerruti, Jacob T. Jackson, and Joseph A. Trapani
- Subjects
CD4-Positive T-Lymphocytes ,Lung Neoplasms ,CD3 Complex ,Receptor, ErbB-2 ,medicine.medical_treatment ,Immunoglobulin Variable Region ,Mice, SCID ,Lymphocyte Activation ,Biochemistry ,Immunotherapy, Adoptive ,Jurkat Cells ,Mice ,Chlorocebus aethiops ,Tumor Cells, Cultured ,Cytotoxic T cell ,Immunoglobulin Fragments ,Mice, Knockout ,Mice, Inbred BALB C ,Membrane Glycoproteins ,CD28 ,Antibodies, Monoclonal ,Hematology ,3T3 Cells ,Recombinant Proteins ,medicine.anatomical_structure ,COS Cells ,Sarcoma, Experimental ,Colorectal Neoplasms ,Pore Forming Cytotoxic Proteins ,T cell ,Recombinant Fusion Proteins ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Breast Neoplasms ,Biology ,Adenocarcinoma ,Major histocompatibility complex ,Transfection ,Interferon-gamma ,Antigen ,CD28 Antigens ,Antigens, Neoplasm ,medicine ,Animals ,Humans ,Perforin ,T-cell receptor ,Membrane Proteins ,Cell Biology ,Immunotherapy ,Neoplasms, Experimental ,Xenograft Model Antitumor Assays ,Tumor Escape ,Cancer research ,biology.protein ,Spleen ,T-Lymphocytes, Cytotoxic - Abstract
Tumor cells are usually weakly immunogenic as they largely express self-antigens and can down-regulate major histocompatability complex/peptide molecules and critical costimulatory ligands. The challenge for immunotherapies has been to provide vigorous immune effector cells that circumvent these tumor escape mechanisms and eradicate established tumors. One promising approach is to engineer T cells with single-chain antibody receptors, and since T cells require 2 distinct signals for optimal activation, we have compared the therapeutic efficacy of erbB2-reactive chimeric receptors that contain either T-cell receptor zeta (TCR-ζ) or CD28/TCR-ζ signaling domains. We have demonstrated that primary mouse CD8+ T lymphocytes expressing the single-chain Fv (scFv)–CD28-ζ receptor have a greater capacity to secrete Tc1 cytokines, induce T-cell proliferation, and inhibit established tumor growth and metastases in vivo. The suppression of established tumor burden by cytotoxic T cells expressing the CD28/TCR-ζ chimera was critically dependent upon their interferon gamma (IFN-γ) secretion. Our study has illustrated the practical advantage of engineering a T-cell signaling complex that codelivers CD28 activation, dependent only upon the tumor's expression of the appropriate tumor associated antigen.
- Published
- 2002
49. Declined Presentation
- Author
-
Don Metcalf, Benjamin J. Shields, Matthew P. McCormack, Chayanica Nasa, Clifford W. Bogue, Warren S. Alexander, and Jacob T. Jackson
- Subjects
Cancer Research ,media_common.quotation_subject ,Cell Biology ,Hematology ,Biology ,Self renewal ,Presentation ,Genetics ,Cancer research ,Homeobox ,Stem cell ,Molecular Biology ,Transcription factor ,media_common - Published
- 2014
- Full Text
- View/download PDF
50. Drosophila myb is required for the G2/M transition and maintenance of diploidy
- Author
-
Siau Min Fung, Alisa L. Katzen, Harmon Bp, J M Bishop, Jacob T. Jackson, and G. Ramsay
- Subjects
DNA Replication ,G2 Phase ,animal structures ,Cell division ,Cell Survival ,Mutant ,DNA Mutational Analysis ,Molecular Sequence Data ,Mitosis ,Polyploidy ,Proto-Oncogene Proteins c-myb ,Proto-Oncogene Proteins ,Genetics ,Endoreduplication ,Animals ,Wings, Animal ,MYB ,Amino Acid Sequence ,Cell Nucleus ,biology ,Sequence Homology, Amino Acid ,fungi ,Cell Cycle ,Temperature ,biology.organism_classification ,Diploidy ,Drosophila melanogaster ,Phenotype ,Mutation ,Trans-Activators ,Ectopic expression ,Cell Division ,Developmental Biology ,Research Paper - Abstract
The myb proto-oncogenes are thought to have a role in the cell division cycle. We have examined this possibility by genetic analysis in Drosophila melanogaster, which possesses a single myb gene. We have described previously two temperature-sensitive, recessive lethal mutants in Drosophila myb (Dm myb). The phenotypes of these mutants revealed a requirement for myb in diverse cellular lineages throughout the course of Drosophila development. We now report a cellular explanation for these findings by showing that Dm myb is required for both mitosis and prevention of endoreduplication in wing cells. Myb apparently acts at or near the time of the G2/M transition. The two mutant alleles of Dm myb produce the same cellular phenotype, although the responsible mutations are located in different functional domains of the gene product. The mutant phenotype can be partially suppressed by ectopic expression of either cdc2 or string, two genes that are known to promote the transition from G2 to M. We conclude that Dm myb is required for completion of cell division and may serve two independent functions: promotion of mitosis, on the one hand, and prevention of endoreduplication when cells are arrested in G2, on the other.
- Published
- 1998
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.