51
|
Afar DE, Bhaskar V, Ibsen E, Breinberg D, Henshall SM, Kench JG, Drobnjak M, Powers R, Wong M, Evangelista F, O'Hara C, Powers D, DuBridge RB, Caras I, Winter R, Anderson T, Solvason N, Stricker PD, Cordon-Cardo C, Scher HI, Grygiel JJ, Sutherland RL, Murray R, Ramakrishnan V, Law DA. Preclinical validation of anti-TMEFF2-auristatin E–conjugated antibodies in the treatment of prostate cancer. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.921.3.8] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Current treatments for advanced stage, hormone-resistant prostate cancer are largely ineffective, leading to high patient mortality and morbidity. To fulfill this unmet medical need, we used global gene expression profiling to identify new potential antibody-drug conjugate (ADC) targets that showed maximal prostate cancer-specific expression. TMEFF2, a gene encoding a plasma membrane protein with two follistatin-like domains and one epidermal growth factor–like domain, had limited normal tissue distribution and was highly overexpressed in prostate cancer. Immunohistochemistry analysis using a specific monoclonal antibody (mAb) to human TMEFF2 showed significant protein expression in 74% of primary prostate cancers and 42% of metastatic lesions from lymph nodes and bone that represented both hormone-naïve and hormone-resistant disease. To evaluate anti-TMEFF2 mAbs as potential ADCs, one mAb was conjugated to the cytotoxic agent auristatin E via a cathepsin B–sensitive valine-citrulline linker. This ADC, Pr1-vcMMAE, was used to treat male severe combined immunodeficient mice bearing xenografted LNCaP and CWR22 prostate cancers expressing TMEFF2. Doses of 3 to 10 mg/kg of this specific ADC resulted in significant and sustained tumor growth inhibition, whereas an isotype control ADC had no significant effect. Similar efficacy and specificity was shown with huPr1-vcMMAE, a humanized anti-TMEFF2 ADC. No overt in vivo toxicity was observed with either murine or human ADC, despite significant cross-reactivity of anti-TMEFF2 mAb with the murine TMEFF2 protein, implying minimal toxicity to other body tissues. These data support the further evaluation and clinical testing of huPr1-vcMMAE as a novel therapeutic for the treatment of metastatic and hormone-resistant prostate cancer.
Collapse
Affiliation(s)
| | | | - Eric Ibsen
- 1Protein Design Labs, Inc., Fremont, California
| | | | | | - James G. Kench
- 2Garvan Institute of Medical Research and Departments of
- 5Department of Tissue Pathology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Marija Drobnjak
- 6Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Rick Powers
- 1Protein Design Labs, Inc., Fremont, California
| | | | | | | | | | | | | | - Ruth Winter
- 7Eos Biotechnology, Inc., Fremont, California
| | | | | | | | | | - Howard I. Scher
- 6Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - John J. Grygiel
- 4Medical Oncology, St. Vincent's Hospital, Darlinghurst, Sydney, New South Wales, Australia
| | | | | | | | | |
Collapse
|
52
|
Matsuyama S, Aihara K, Nishino N, Takeda S, Tanizawa K, Kuroda S, Horie M. Enhanced long-term potentiation in vivo in dentate gyrus of NELL2-deficient mice. Neuroreport 2004; 15:417-20. [PMID: 15094495 DOI: 10.1097/00001756-200403010-00007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
NELL2 is a neuron-specific thrombospondin-1-like extracellular protein containing six epidermal growth factor-like domains and is highly expressed in the hippocampus. We have previously shown that NELL2 promotes survival of neurons through mitogen-activated protein kinases. To clarify the function of NELL2 in vivo, we have generated a novel strain of mice with a targeted mutation in the NELL2 gene and assessed long-term potentiation (LTP) in vivo in the dentate gyrus of NELL2-deficient mice using extracellular recording techniques. Production of LTP at perforant path-granule cell synapses was significantly larger in NELL2-deficient mice than in wild-type controls. Thus, we propose that NELL2 plays an important role as a novel suppressor in LTP in vivo in the mouse dentate gyrus.
Collapse
Affiliation(s)
- Shogo Matsuyama
- Division of Molecular Pharmacology and Pharmacogenomics, Department of Genome Sciences, Kobe University Graduate School of Medicine, Hyogo 650-0017, Japan
| | | | | | | | | | | | | |
Collapse
|
53
|
Harms PW, Chang C. Tomoregulin-1 (TMEFF1) inhibits nodal signaling through direct binding to the nodal coreceptor Cripto. Genes Dev 2003; 17:2624-9. [PMID: 14563676 PMCID: PMC280611 DOI: 10.1101/gad.1127703] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Transforming growth factor beta (TGF-beta) signals regulate multiple processes during development and in adult. We recently showed that tomoregulin-1 (TMEFF1), a transmembrane protein, selectively inhibits nodal but not activin in early Xenopus embryos. Here we report that TMEFF1 binds to the nodal coreceptor Cripto, but does not associate with either nodal or the type I ALK (activin receptor-like kinase) 4 receptor in coimmunoprecipitation assays. The inhibition of the nodal signaling by TMEFF1 in Xenopus ectodermal explants is rescued with wild-type but not mutant forms of Cripto. Furthermore, we show that the Cripto-FRL1-Cryptic (CFC) domain in Cripto, which is essential for its binding to ALK4, is also important for its interaction with TMEFF1. Our results demonstrate for the first time that nodal signaling can be regulated by a novel mechanism of blocking the Cripto coreceptor.
Collapse
Affiliation(s)
- Paul W Harms
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | |
Collapse
|
54
|
Aihara K, Kuroda S, Kanayama N, Matsuyama S, Tanizawa K, Horie M. A neuron-specific EGF family protein, NELL2, promotes survival of neurons through mitogen-activated protein kinases. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2003; 116:86-93. [PMID: 12941464 DOI: 10.1016/s0169-328x(03)00256-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
NELL2 is a neuron-specific thrombospondin-1-like extracellular protein containing six epidermal growth factor-like domains. NELL2 is highly expressed in the hippocampus and cerebral cortex. Although the involvement of NELL2 in neural functions has been inferred from its expression and biochemical profiles, biological roles of NELL2 remain uncertain. We evaluated the survival effect of NELL2 using primary cultured neurons from fetal rat brain following treatment with a recombinant NELL2 protein. NELL2 increased survival of neurons from the hippocampus and cerebral cortex. We further examined the protective effect of NELL2 from oxygen-glucose deprivation- and beta-amyloid-induced neuronal death, and found that NELL2 did not protect neurons from these insults. To understand signaling properties underlying the survival effect, we studied activation of mitogen-activated protein kinases (MAPKs) by NELL2. Treatment of primary cultured cells from the hippocampus with NELL2 enhanced phosphorylation of c-jun N-terminal kinase (JNK), whereas phosphorylation of extracellular signal-regulated kinase (ERK) was decreased by NELL2 treatment. NELL2-enhanced survival of hippocampal neurons was completely blocked by SP600125, an anthrapyrazolone inhibitor of JNK, while treatment of MEK (MAPK/ERK kinase) inhibitors per se enhanced survival of neurons similar to NELL2 treatment. These results suggest that NELL2 promotes survival of neurons by modulating MAPK activities.
Collapse
Affiliation(s)
- Koutoku Aihara
- Second Institute of New Drug Discovery, Otsuka Pharmaceutical Co Ltd, 463-10 Kagasuno, Kawauchi-cho, Tokushima 771-0192, Japan
| | | | | | | | | | | |
Collapse
|
55
|
Vannahme C, Gösling S, Paulsson M, Maurer P, Hartmann U. Characterization of SMOC-2, a modular extracellular calcium-binding protein. Biochem J 2003; 373:805-14. [PMID: 12741954 PMCID: PMC1223551 DOI: 10.1042/bj20030532] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2003] [Accepted: 05/13/2003] [Indexed: 01/11/2023]
Abstract
We have isolated the novel gene SMOC-2, which encodes a secreted modular protein containing an EF-hand calcium-binding domain homologous to that in BM-40. It further consists of two thyroglobulin-like domains, a follistatin-like domain and a novel domain found only in the homologous SMOC-1. Phylogenetic analysis of the calcium-binding domain sequences showed that SMOC-1 and -2 form a separate group within the BM-40 family. The human and mouse SMOC-2 sequences are coded for by genes consisting of 13 exons located on chromosomes 6 and 17, respectively. Analysis of recombinantly expressed protein showed that SMOC-2 is a glycoprotein with a calcium-dependent conformation. Results from Northern blots and reverse transcription PCR revealed a widespread expression in many tissues.
Collapse
Affiliation(s)
- Christian Vannahme
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann Strasse 52, D-50931 Cologne, Germany
| | | | | | | | | |
Collapse
|
56
|
Abstract
TMEFF2 is a novel transmembrane protein, containing two follistatin domains and an epidermal growth factor-like motif that is mainly expressed in the prostate and brain. Recently, we showed that expression of TMEFF2 could inhibit prostate cancer cell growth. In addition, the TMEFF2 gene is frequently hypermethylated in human tumor cells, suggesting that it might be a tumor suppressor gene. We cloned the 5'-flanking region of the human TMEFF2 gene and using a luciferase reporter assay showed that it contains a functional promoter. The 0.7 kb region upstream to the TMEFF2 transcription start site encompasses the minimal promoter required for TMEFF2 expression. Sequence analysis of the TMEFF2 promoter revealed potential binding sites for several transcription factors including Sp1 and an E-box that could be recognized by c-Myc. An inverse correlation between TMEFF2 and c-Myc expression was found in CWR22 prostate xenografts. Reporter gene and mobility shift assays demonstrated that c-Myc could repress TMEFF2 gene expression through its cognate site. In light of the probable role of TMEFF2 in inhibiting cell growth, its suppression may contribute to the oncogenic properties of c-Myc.
Collapse
Affiliation(s)
- Sigal Gery
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, CA 90048, USA.
| | | |
Collapse
|
57
|
Abstract
TMEFF1 is a novel transmembrane protein, containing two follisatin domains and an epidermal growth factor-like region. These structural domains suggest a role for TMEFF1 in growth factor signaling. TMEFF1 fused to enhanced green fluorescent protein revealed that TMEFF1 is expressed on the cell membrane. Northern analysis of normal human tissue showed that TMEFF1 is predominantly expressed in the brain. Study of cancer cell lines from different tissues including the brain, demonstrated moderate to low levels of TMEFF1 in most of these transformed cell lines. Furthermore, quantitative real-time RT-PCR analysis of 54 brain tumors showed that most of these tumors (96%) had lower levels of TMEFF1 expression than normal brain tissue. Interestingly, ectopic expression of TMEFF1 in brain cancer cells resulted in their growth inhibition. These data suggest that TMEFF1 may behave as a tumor suppressor gene in brain cancers.
Collapse
Affiliation(s)
- Sigal Gery
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | |
Collapse
|
58
|
Chang C, Eggen BJL, Weinstein DC, Brivanlou AH. Regulation of nodal and BMP signaling by tomoregulin-1 (X7365) through novel mechanisms. Dev Biol 2003; 255:1-11. [PMID: 12618130 DOI: 10.1016/s0012-1606(02)00075-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
During early vertebrate development, members of the transforming growth factor beta (TGFbeta) family play important roles in a variety of processes, including germ layer specification, patterning, cell differentiation, migration, and organogenesis. The activities of TGFbetas need to be tightly controlled to ensure their function at the right time and place. Despite identification of multiple regulators of Bone Morphogenetic Protein (BMP) subfamily ligands, modulators of the activin/nodal class of TGFbeta ligands are limited, and include follistatin, Cerberus, and Lefty. Recently, a membrane protein, tomoregulin-1 (TMEFF1, originally named X7365), was isolated and found to contain two follistatin modules in addition to an Epidermal Growth Factor (EGF) domain, suggesting that TMEFF1 may participate in regulation of TGFbeta function. Here, we show that, unlike follistatin and follistatin-related gene (FLRG), TMEFF1 inhibits nodal but not activin in Xenopus. Interestingly, both the follistatin modules and the EGF motif contribute to nodal inhibition. A soluble protein containing the follistatin and the EGF domains, however, is not sufficient for nodal inhibition; the location of TMEFF1 at the membrane is essential for its function. These results suggest that TMEFF1 inhibits nodal through a novel mechanism. TMEFF1 also blocks mesodermal, but not epidermal induction by BMP2. Unlike nodal inhibition, regulation of BMP activities by TMEFF1 requires the latter's cytoplasmic tail, while deletion of either the follistatin modules or the EGF motif does not interfere with the BMP inhibitory function of TMEFF1. These results imply that TMEFF1 may employ different mechanisms in the regulation of nodal and BMP signals. In Xenopus, TMEFF1 is expressed from midgastrula stages onward and is enriched in neural tissue derivatives. This expression pattern suggests that TMEFF1 may modulate nodal and BMP activities during neural patterning. In summary, our data demonstrate that tomoregulin-1 is a novel regulator of nodal and BMP signaling during early vertebrate embryogenesis.
Collapse
Affiliation(s)
- Chenbei Chang
- Laboratory of Vertebrate Molecular Embryology, The Rockefeller University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
59
|
Perkins D, Pereira EFR, Aurelian L. The herpes simplex virus type 2 R1 protein kinase (ICP10 PK) functions as a dominant regulator of apoptosis in hippocampal neurons involving activation of the ERK survival pathway and upregulation of the antiapoptotic protein Bag-1. J Virol 2003; 77:1292-305. [PMID: 12502846 PMCID: PMC140789 DOI: 10.1128/jvi.77.2.1292-1305.2003] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Herpes simplex virus types 1 and 2 (HSV-1 and HSV-2) can trigger or block apoptosis in a cell type-dependent manner. We have recently shown that the protein kinase activity of the large subunit of the HSV-2 ribonucleotide reductase (R1) protein (ICP10 PK) blocks apoptosis in cultured hippocampal neurons by activating the extracellular signal-regulated kinase (ERK) survival pathway (Perkins et al., J. Virol. 76:1435-1449, 2002). The present studies were designed to better elucidate the mechanism of ICP10 PK-induced neuroprotection and determine whether HSV-1 has similar activity. The data indicate that apoptosis inhibition by ICP10 PK involves a c-Raf-1-dependent mechanism and induction of the antiapoptotic protein Bag-1 by the activated ERK survival pathway. Also associated with neuroprotection by ICP10 PK are increased activation/stability of the transcription factor CREB and stabilization of the antiapoptotic protein Bcl-2. HSV-1 and the ICP10 PK-deleted HSV-2 mutant ICP10DeltaPK activate JNK, c-Jun, and ATF-2, induce the proapoptotic protein BAD, and trigger apoptosis in hippocampal neurons. c-Jun activation and apoptosis are inhibited in hippocampal cultures infected with HSV-1 in the presence of the JNK inhibitor SP600125, suggesting that JNK/c-Jun activation is required for HSV-1-induced apoptosis. Ectopically delivered ICP10 PK (but not its PK-negative mutant p139) inhibits apoptosis triggered by HSV-1 or ICP10DeltaPK. Collectively, the data indicate that ICP10 PK-induced activation of the ERK survival pathway results in Bag-1 upregulation and overrides the proapoptotic JNK/c-Jun signal induced by other viral proteins.
Collapse
Affiliation(s)
- D Perkins
- Departments of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | |
Collapse
|
60
|
Vannahme C, Smyth N, Miosge N, Gösling S, Frie C, Paulsson M, Maurer P, Hartmann U. Characterization of SMOC-1, a novel modular calcium-binding protein in basement membranes. J Biol Chem 2002; 277:37977-86. [PMID: 12130637 DOI: 10.1074/jbc.m203830200] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have isolated the novel gene SMOC-1 that encodes a secreted modular protein containing an EF-hand calcium-binding domain homologous to that in BM-40. It further consists of two thyroglobulin-like domains, a follistatin-like domain and a novel domain. Recombinant expression in human cells showed that SMOC-1 is a glycoprotein with a calcium-dependent conformation. Results from Northern blots, reverse transcriptase-PCR, and immunoblots revealed a widespread expression in many tissues. Immunofluorescence studies with an antiserum directed against recombinant human SMOC-1 demonstrated a basement membrane localization of the protein and additionally its presence in other extracellular matrices. Immunogold electron microscopy confirmed the localization of SMOC-1 within basement membranes in kidney and skeletal muscle as well as its expression in the zona pellucida surrounding the oocyte.
Collapse
Affiliation(s)
- Christian Vannahme
- Institute for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann Strasse 52, D-50931 Cologne, Germany
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Gery S, Sawyers CL, Agus DB, Said JW, Koeffler HP. TMEFF2 is an androgen-regulated gene exhibiting antiproliferative effects in prostate cancer cells. Oncogene 2002; 21:4739-46. [PMID: 12101412 DOI: 10.1038/sj.onc.1205142] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2001] [Revised: 10/23/2001] [Accepted: 10/31/2001] [Indexed: 11/09/2022]
Abstract
We have identified a gene that is highly expressed in the androgen-dependent prostate cancer cell line, LNCaP. Sequence analysis revealed that it was identical to a recently cloned gene designated TMEFF2, which encodes a transmembrane protein containing an epidermal growth factor (EGF)-like motif and two follistatin domains. This gene was highly expressed only in primary samples of normal prostate and prostate cancer as well as normal brain. Expression of the gene was controlled by androgen as shown by dihydrotestosterone markedly increasing TMEFF2 expression in LNCaP cells. Also, androgen-dependent human prostate cancer xenografts (CWR22) expressed high levels of TMEFF2 and these levels markedly decreased by day 10 after castration of the mice. Furthermore, a large number of androgen-dependent xenografts (CWR22, LuCaP-35, LAPC-4AD, LAPC-9AD) exhibited higher levels of TMEFF2 mRNA than androgen-independent xenografts (CWR22R, LAPC-3AI, LAPC-4AI, LAPC-9AI). Ectopic expression of TMEFF2 in DU145 and PC3 cells resulted in their prominent inhibition of growth. Taken together, the results demonstrate that TMEFF2 is a androgen-regulated gene, which can suppress growth of prostate cancer cells and our xenograft data show that escape of prostate cancer cells from androgen modulation causes them to decrease their expression of this gene, which may result in their more malignant behavior.
Collapse
Affiliation(s)
- Sigal Gery
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles, California, CA 90048, USA.
| | | | | | | | | |
Collapse
|
62
|
Siegel DA, Huang MK, Becker SF. Ectopic dendrite initiation: CNS pathogenesis as a model of CNS development. Int J Dev Neurosci 2002; 20:373-89. [PMID: 12175877 DOI: 10.1016/s0736-5748(02)00055-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neuronal storage diseases are a rare group of disorders with profound clinical consequences including severe mental retardation and death in early childhood. A subset of these disorders, those with elevated levels of GM2 ganglioside, are further characterized by the reinitiation of primary dendrites on mature cortical neurons. These ectopic dendrites are unusual as primary dendrite initiation is normally confined to a narrow developmental window. Thus, ectopic dendritogenesis appears to be a recapitulation of the normal developmental program temporally displaced. Consequently, understanding ectopic dendritogenesis should offer insights into both the pathogenesis of the neuronal storage diseases as well as mechanisms of normal CNS development. Using a feline model of GM2 gangliosidosis, we compared patterns of gene expression in normal newborn and mature diseased animals (both undergoing active primary dendritogenesis) with normal, mature controls (where primary dendritogenesis has ceased). From this work, we have identified two genes that appear to function in primary dendrite initiation. One, tomoregulin, is an integral membrane protein with both EGF- and follistatin-like motifs in its extracellular domain. The second, Tristanin, is a member of the positive regulatory domain (PRD) family of a zinc-finger transcription factors. Both genes are up regulated in the disease state, and both show a shift in their intracellular location to the nucleus in diseased animals that is not observed in age matched controls. In normal mouse brain, tomoregulin and Tristanin reveal developmental patterns consistent with a role in dendrite initiation and show changes in subcellular localization similar to that observed in the cat.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cats
- Cell Differentiation/genetics
- Cells, Cultured
- Cerebral Cortex/abnormalities
- Cerebral Cortex/growth & development
- Cerebral Cortex/pathology
- DNA, Complementary/analysis
- DNA, Complementary/genetics
- DNA-Binding Proteins
- Dendrites/pathology
- Disease Models, Animal
- Fetus
- Gangliosidoses, GM2/genetics
- Gangliosidoses, GM2/pathology
- Gangliosidoses, GM2/physiopathology
- Genetic Testing
- Immunohistochemistry
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Molecular Sequence Data
- Neoplasm Proteins
- Pyramidal Cells/abnormalities
- Pyramidal Cells/pathology
- RNA, Messenger/analysis
- RNA, Messenger/genetics
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- Transcription Factors/genetics
- Transcription Factors/isolation & purification
Collapse
Affiliation(s)
- Donald A Siegel
- Department of Neuroscience, Albert Einstein College of Medicine, Kennedy Center, Bronx, NY 10461, USA.
| | | | | |
Collapse
|
63
|
Glynne-Jones E, Harper ME, Seery LT, James R, Anglin I, Morgan HE, Taylor KM, Gee JM, Nicholson RI. TENB2, a proteoglycan identified in prostate cancer that is associated with disease progression and androgen independence. Int J Cancer 2001; 94:178-84. [PMID: 11668495 DOI: 10.1002/ijc.1450] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
TENB2 encodes a putative transmembrane proteoglycan, related to the EGF/heregulin family of growth factors and follistatin, which has been identified through the application of a differential display technique to a xenograft model of prostate cancer. Northern analysis and competitive PCR were used to demonstrate significantly increased TENB2 expression (p = 0.0003) on the acquisition of androgen independence in the model system. TENB2 is also overexpressed in clinical prostate carcinoma vs. its benign counterpart (p < 0.0001), with particular prominence in high-grade tumours, and shows a high degree of tissue specificity, being detected on a multitissue Northern array exclusively in brain and prostate material. Studies of recombinant protein expression demonstrate that TENB2 is a chondroitin sulphate proteoglycan. The presence of an EGF and 2 follistatin domains suggests a role in the regulation of growth factor signalling either as a ligand precursor, a membrane-bound receptor or as a binding protein for growth factors. These data are indicative of a significant role for TENB2 in the progression of poorly differentiated tumour types, with implications for prostate cancer detection, prognosis and therapy.
Collapse
Affiliation(s)
- E Glynne-Jones
- Tenovus Cancer Research Centre, Welsh School of Pharmacy, Cardiff University, Wales, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Kanemoto N, Horie M, Omori K, Nishino N, Kondo M, Noguchi K, Tanigami A. Expression of TMEFF1 mRNA in the mouse central nervous system: precise examination and comparative studies of TMEFF1 and TMEFF2. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 86:48-55. [PMID: 11165370 DOI: 10.1016/s0169-328x(00)00257-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
TMEFF1 and TMEFF2 are putative transmembrane proteins comprised of one epidermal growth factor (EGF)-like domain and two follistatin-like domains. Both TMEFF1 and TMEFF2 are predominantly expressed in the brain. We previously demonstrated that recombinant TMEFF2 protein can promote survival of neurons in primary culture and determined expression sites of TMEFF2 mRNA in the mouse central nervous system. To extend our understanding of TMEFF protein functions, we compared precise sites of expression of TMEFF1 and TMEFF2 mRNA using in situ hybridization analysis. Although both TMEFF genes are widely expressed in the brain, they exhibit different patterns of expression. TMEFF1 showed comparatively higher signals in the pyramidal cells of fifth layer of the cerebral neocortex, CA3, CA1 and subiculum regions of the hippocampus, locus coeruleus, and dentate cerebellar nucleus. In contrast, TMEFF2 is highly expressed in the medial habenular, CA2, CA3 and dentate gyrus region of the hippocampus, corpus callosum, cerebellar cortex and cranial nerve nuclei (III, IV, VII, X, XII). The results presented here indicate that expression of TMEFF1 and TMEFF2 are regulated differently and that they play region-specific roles in the central nervous system.
Collapse
Affiliation(s)
- N Kanemoto
- Otsuka GEN Research Institute, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | | | | | | | | | | | | |
Collapse
|