84 results on '"Lamothe B"'
Search Results
2. POS0211 PREDICTORS OF PROGRESSION AND MORTALITY IN PATIENTS WITH PREVALENT RHEUMATOID ARTHRITIS AND INTERSTITIAL LUNG DISEASE: A PROSPECTIVE COHORT STUDY
- Author
-
Mena-Vázquez, N., primary, Rojas-Giménez, M., additional, Romero-Barco, C. M., additional, Manrique Arija, S., additional, Espildora, F., additional, Aguilar-Hurtado, M. C., additional, Ortega Castro, R., additional, Añón Oñate, I., additional, Pérez Albaladejo, L., additional, Godoy-Navarrete, F., additional, Ureña, I., additional, Velloso Feijoo, M., additional, Redondo, R., additional, Jiménez-Núñez, F. G., additional, Panero Lamothe, B., additional, Padin-Martín, M. I., additional, and Fernandez-Nebro, A., additional
- Published
- 2021
- Full Text
- View/download PDF
3. Genetic Approaches to the Mechanisms and Biology of Signal Transduction by Insulin and IGF-I Receptors.
- Author
-
Lamothe, B., primary, Baudry, A., additional, Bucchini, D., additional, Jami, J., additional, and Joshi, R.L., additional
- Published
- 1998
- Full Text
- View/download PDF
4. IGF-1 receptor as an alternative receptor for metabolic signaling in insulin receptor-deficient muscle cells
- Author
-
Baudry, A., Lamothe, B., Bucchini, D., Jami, J., Montarras, D., Pinset, C., and Joshi, R.L.
- Published
- 2001
- Full Text
- View/download PDF
5. Pharmacodynamics and proteomic analysis of acalabrutinib therapy: similarity of on-target effects to ibrutinib and rationale for combination therapy
- Author
-
Patel, V K, primary, Lamothe, B, additional, Ayres, M L, additional, Gay, J, additional, Cheung, J P, additional, Balakrishnan, K, additional, Ivan, C, additional, Morse, J, additional, Nelson, M, additional, Keating, M J, additional, Wierda, W G, additional, Marszalek, J R, additional, and Gandhi, V, additional
- Published
- 2017
- Full Text
- View/download PDF
6. AB0361 Body composition and physical activity in patients with rheumatoid arthritis (AR)
- Author
-
Manrique-Arija, S, primary, Mena-Vazquez, N, additional, Rojas-Giménez, M, additional, Fuego, C, additional, Abad-Sanchez, S, additional, Ureña-Garnica, I, additional, Domic, C, additional, Jiménez-Núñez, FG, additional, Ordoñez-Cañizares, MC, additional, Caparros-Ruiz, R, additional, Cano-Garcia, L, additional, Belmonte, A, additional, Panero-Lamothe, B, additional, and Fernandez-Nebro, A, additional
- Published
- 2017
- Full Text
- View/download PDF
7. Pharmacodynamics and proteomic analysis of acalabrutinib therapy: similarity of on-target effects to ibrutinib and rationale for combination therapy
- Author
-
Patel, V K, Lamothe, B, Ayres, M L, Gay, J, Cheung, J P, Balakrishnan, K, Ivan, C, Morse, J, Nelson, M, Keating, M J, Wierda, W G, Marszalek, J R, and Gandhi, V
- Abstract
Acalabrutinib, a highly selective Bruton’s tyrosine kinase inhibitor, is associated with high overall response rates and durable remission in previously treated chronic lymphocytic leukemia (CLL); however, complete remissions were limited. To elucidate on-target and pharmacodynamic effects of acalabrutinib, we evaluated several laboratory endpoints, including proteomic changes, chemokine modulation and impact on cell migration. Pharmacological profiling of samples from acalabrutinib-treated CLL patients was used to identify strategies for achieving deeper responses, and to identify additive/synergistic combination regimens. Peripheral blood samples from 21 patients with relapsed/refractory CLL in acalabrutinib phase I (100–400 mg/day) and II (100 mg BID) clinical trials were collected prior to and on days 8 and 28 after treatment initiation and evaluated for plasma chemokines, reverse phase protein array, immunoblotting and pseudoemperipolesis. The on-target pharmacodynamic profile of acalabrutinib in CLL lymphocytes was comparable to ibrutinib in measures of acalabrutinib-mediated changes in CCL3/CCL4 chemokine production, migration assays and changes in B-cell receptor signaling pathway proteins and other downstream survival proteins. Among several CLL-targeted agents, venetoclax, when combined with acalabrutinib, showed optimal complementary activity in vitro, ex vivo and in vivo in TCL-1 adoptive transfer mouse model system of CLL. These findings support selective targeting and combinatorial potential of acalabrutinib.
- Published
- 2018
- Full Text
- View/download PDF
8. Distinct molecular mechanism for initiating TRAF6 signalling
- Author
-
Ye H., Arron J.R., Lamothe B., Cirilli M., Kobayashi T., Shevde N.K., Segal D., Dzivenu O.K., Volgodskaia M., Yim M., Du K., Singh S., Pike J.W., Darnay B.G., Choi Y., and Wu H.
- Subjects
Biologia Strutturale ,Cristallografia ,TNF ,Recettori cellulari - Abstract
Tumour-necrosis factor (TNF) receptor-associated factor 6 (TRAF6) is the only TRAF family member that participates in signal transduction of both the TNF receptor (TNFR) superfamily and the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) superfamily; it is important for adaptive immunity, innate immunity and bone homeostasis. Here we report crystal structures of TRAF6, alone and in complex with TRAF6-binding peptides from CD40 and TRANCE-R (also known as RANK), members of the TNFR superfamily, to gain insight into the mechanism by which TRAF6 mediates several signalling cascades. A 40 degrees difference in the directions of the bound peptides in TRAF6 and TRAF2 shows that there are marked structural differences between receptor recognition by TRAF6 and other TRAFs. The structural determinant of the petide TRAF6 interaction reveals a Pro-X-Glu-X-X-(aromatic/acidic residue) TRAF6-binding motif, which is present not only in CD40 and TRANCE-R but also in the three IRAK adapter kinases for IL-1R/TLR signalling. Cell-permeable peptides with the TRAF6-binding motif inhibit TRAF6 signalling, which indicates their potential as therapeutic modulators. Our studies identify a universal mechanism by which TRAF6 regulates several signalling cascades in adaptive immunity, innate immunity and bone homeostasis.
- Published
- 2002
9. Genetic manipulation of insulin action and beta-cell function in mice
- Author
-
Lamothe, B., Duvillié, B., Cordonnier, N, Baudry, A, Bucchini, D, Jami, J, Joshi, Rajiv L, Saint-Just, S., Joshi, Rajiv L., Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS Paris), and Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Mice, Knockout ,MESH: Humans ,MESH: Mice, Transgenic ,MESH: Islets of Langerhans ,[SDV]Life Sciences [q-bio] ,Mice, Transgenic ,MESH: Insulin ,MESH: Mice, Knockout ,[SDV] Life Sciences [q-bio] ,Disease Models, Animal ,Islets of Langerhans ,Mice ,Diabetes Mellitus, Type 2 ,Animals ,Humans ,Insulin ,MESH: Animals ,MESH: Disease Models, Animal ,MESH: Mice ,MESH: Diabetes Mellitus, Type 2 - Abstract
International audience; Transgenic and gene targeting approaches have now been applied to a number of genes in order to investigate the metabolic disorders that would result by manipulating insulin action or pancreatic beta-cell function in the mouse. The availability of such mutant mice will allow in the future to develop animal models in which the pathophysiologies resulting from polygenic defects might be reconstituted and studied in detail. Such animal models hopefully will lead to better understanding of complex polygenic diseases such as non-insulin-dependent diabetes mellitus (NIDDM).
- Published
- 1998
10. Genetically engineered mice as animal models for NIDDM
- Author
-
Joshi, Rajiv L, Joshi, J, Lamothe, B, Bucchini, D, Jami, J, Centre interdisciplinaire de recherche en biologie (CIRB), Labex MemoLife, École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Collège de France (CdF (institution))-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and Joshi, Rajiv L.
- Subjects
Genetically modified mouse ,MESH: Mice, Transgenic ,Transgene ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,Biophysics ,Mice, Transgenic ,MESH: Insulin ,MESH: Gene Targeting ,Biochemistry ,Genome ,Islets of Langerhans ,Mice ,Insulin resistance ,Transgenic mouse ,Structural Biology ,Genetics ,medicine ,Animals ,Humans ,Insulin ,MESH: Animals ,Molecular Biology ,Gene ,MESH: Mice ,Insulin action ,MESH: Humans ,biology ,Insulin secretion ,MESH: Islets of Langerhans ,Diabetes ,Gene targeting ,Cell Biology ,medicine.disease ,[SDV] Life Sciences [q-bio] ,Disease Models, Animal ,Insulin receptor ,Diabetes Mellitus, Type 2 ,Gene Targeting ,biology.protein ,MESH: Genetic Engineering ,MESH: Disease Models, Animal ,Genetic Engineering ,MESH: Diabetes Mellitus, Type 2 - Abstract
International audience; Genetically engineered animals carrying defined alterations in their genome can represent invaluable tools for better understanding complex polygenic diseases such as non-insulin-dependent diabetes mellitus (NIDDM) at the molecular level. The structure or expression of a number of genes potentially involved in insulin action or pancreatic beta-cell function have recently been altered in the mouse using transgenic or gene-targeting approaches. The obtention of such mice is the first step towards the development of animal models carrying multiple gene defects which would be very useful in NIDDM research.
- Published
- 1997
11. Partial rescue of insulin receptor-deficient mice by transgenic complementation with an activated insulin receptor in the liver
- Author
-
Baudry, A., Jackerott, Malene, Lamothe, B., Kozyrev, S.V., Leroux, L., Durel, B., Saint-Just, S., Joshi, R.L., Baudry, A., Jackerott, Malene, Lamothe, B., Kozyrev, S.V., Leroux, L., Durel, B., Saint-Just, S., and Joshi, R.L.
- Published
- 2002
12. Crystal structure of the N-terminal domain of TRAF6
- Author
-
Yin, Q., primary, Lin, S.-C., additional, Lamothe, B., additional, Lu, M., additional, Lo, Y.-C., additional, Hura, G., additional, Zheng, L., additional, Rich, R.L., additional, Campos, A.D., additional, Myszka, D.G., additional, Lenardo, M.J., additional, Darnay, B.G., additional, and Wu, H., additional
- Published
- 2009
- Full Text
- View/download PDF
13. Crystal structure of TRAF6 in complex with Ubc13 in the C2 space group
- Author
-
Yin, Q., primary, Lin, S.-C., additional, Lamothe, B., additional, Lu, M., additional, Lo, Y.-C., additional, Hura, G., additional, Zheng, L., additional, Rich, R.L., additional, Campos, A.D., additional, Myszka, D.G., additional, Lenardo, M.J., additional, Darnay, B.G., additional, and Wu, H., additional
- Published
- 2009
- Full Text
- View/download PDF
14. Crystal structure of TRAF6 in complex with Ubc13 in the P1 space group
- Author
-
Yin, Q., primary, Lin, S.-C., additional, Lamothe, B., additional, Lu, M., additional, Lo, Y.-C., additional, Hura, G., additional, Zheng, L., additional, Rich, R.L., additional, Campos, A.D., additional, Myszka, D.G., additional, Lenardo, M.J., additional, Darnay, B.G., additional, and Wu, H., additional
- Published
- 2009
- Full Text
- View/download PDF
15. Endocrine pancreas in insulin receptor-deficient mouse pups.
- Author
-
Jackerott, M, primary, Baudry, A, additional, Lamothe, B, additional, Bucchini, D, additional, Jami, J, additional, and Joshi, R L, additional
- Published
- 2001
- Full Text
- View/download PDF
16. Insulin receptor‐deficient cells as a new tool for dissecting complex interplay in insulin and insulin‐like growth factors
- Author
-
Lamothe, B, primary, Baudry, A, additional, Christoffersen, C.T, additional, De Meyts, P, additional, Jami, J, additional, Bucchini, D, additional, and Joshi, R.L, additional
- Published
- 1998
- Full Text
- View/download PDF
17. Targeted disruption of the insulin receptor gene in the mouse results in neonatal lethality.
- Author
-
Joshi, R. L., primary, Lamothe, B., additional, Cordonnier, N., additional, Mesbah, K., additional, Monthioux, E., additional, Jami, J., additional, and Bucchini, D., additional
- Published
- 1996
- Full Text
- View/download PDF
18. Interaction of p85 subunit of PI 3-kinase with insulin and IGF-1 receptors analysed by using the two-hybrid system
- Author
-
Lamothe, B., primary, Bucchini, D., additional, Jami, J., additional, and Joshi, R.L., additional
- Published
- 1995
- Full Text
- View/download PDF
19. Association between imatinib-resistant BCR-ABL mutation-negative leukemia and persistent activation of LYN kinase.
- Author
-
Wu J, Meng F, Kong L, Peng Z, Ying Y, Bornmann WG, Darnay BG, Lamothe B, Sun H, Talpaz M, Donato NJ, Wu, Ji, Meng, Feng, Kong, Ling-Yuan, Peng, Zhenghong, Ying, Yunming, Bornmann, William G, Darnay, Bryant G, Lamothe, Betty, and Sun, Hanshi
- Abstract
Background: Imatinib is a tyrosine kinase inhibitor that is used to treat chronic myelogenous leukemia (CML). BCR-ABL mutations are associated with failure of imatinib treatment in many CML patients. LYN kinase regulates survival and responsiveness of CML cells to inhibition of BCR-ABL kinase, and differences in LYN regulation have been found between imatinib-sensitive and -resistant CML cell lines.Methods: We evaluated cells from 12 imatinib-resistant CML patients with mutation-negative BCR-ABL and from six imatinib-sensitive patients who discontinued therapy because of imatinib intolerance. Phosphorylation of BCR-ABL and LYN was assessed in patient cells and cell lines by immunoblotting with activation state-specific antibodies, co-immunoprecipitation studies, and mass spectroscopy analysis of phosphopeptides. Cell viability, caspase activation, and apoptosis were also measured. Mutations were analyzed by sequencing. The effect of silencing LYN with short interfering RNAs (siRNAs) or reducing activation by treatment with tyrosine kinase inhibitors was evaluated in cell lines and patient cells.Results: Imatinib treatment suppressed LYN phosphorylation in cells from imatinib-sensitive CML patients and imatinib-sensitive cell lines. Imatinib treatment blocked BCR-ABL signaling but did not suppress LYN phosphorylation in cells from imatinib-resistant patients, and persistent activation of LYN kinase was not associated with mutations in LYN kinase or its carboxyl-terminal regulatory domains. Unique LYN phosphorylation sites (tyrosine-193 and tyrosine-459) and associated proteins (c-Cbl and p80) were identified in cells from imatinib-resistant patients. Reducing LYN expression (siRNA) or activation (dasatinib) was associated with loss of cell survival and cytogenetic or complete hematologic responses in imatinib-resistant disease.Conclusions: LYN activation was independent of BCR-ABL in cells from imatinib-resistant patients. Thus, LYN kinase may be involved in imatinib resistance in CML patients with mutation-negative BCR-ABL and its direct inhibition is consistent with clinical responses in these patients. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
20. Genetic Approaches to the Mechanisms and Biology of Signal Transduction by Insulin and IGF-I Receptors.
- Author
-
Lamothe, B., Baudry, A., Bucchini, D., Jami, J., and Joshi, R.L.
- Published
- 1998
- Full Text
- View/download PDF
21. Reexamining interaction of the SH2 domains of SYP and GAP with insulin and IGF-1 receptors in the two-hybrid system
- Author
-
Lamothe, B., Bucchini, D., Jami, J., and Joshi, R. L.
- Published
- 1996
- Full Text
- View/download PDF
22. The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway
- Author
-
Lamothe Betty, Mattoon Dawn R, Lax Irit, and Schlessinger Joseph
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Abstract Background Gab1 is a docking protein that recruits phosphatidylinositol-3 kinase (PI-3 kinase) and other effector proteins in response to the activation of many receptor tyrosine kinases (RTKs). As the autophosphorylation sites on EGF-receptor (EGFR) do not include canonical PI-3 kinase binding sites, it is thought that EGF stimulation of PI-3 kinase and its downstream effector Akt is mediated by an indirect mechanism. Results We used fibroblasts isolated from Gab1-/- mouse embryos to explore the mechanism of EGF stimulation of the PI-3 kinase/Akt anti-apoptotic cell signaling pathway. We demonstrate that Gab1 is essential for EGF stimulation of PI-3 kinase and Akt in these cells and that these responses are mediated by complex formation between p85, the regulatory subunit of PI-3 kinase, and three canonical tyrosine phosphorylation sites on Gab1. Furthermore, complex formation between Gab1 and the protein tyrosine phosphatase Shp2 negatively regulates Gab1 mediated PI-3 kinase and Akt activation following EGF-receptor stimulation. We also demonstrate that tyrosine phosphorylation of ErbB3 may lead to recruitment and activation of PI-3 kinase and Akt in Gab1-/- MEFs. Conclusions The primary mechanism of EGF-induced stimulation of the PI-3 kinase/Akt anti-apoptotic pathway occurs via the docking protein Gab1. However, in cells expressing ErbB3, EGF and neuroregulin can stimulate PI-3 kinase and Akt activation in a Gab1-dependent or Gab1-independent manner.
- Published
- 2004
- Full Text
- View/download PDF
23. Predictors of Progression and Mortality in Patients with Prevalent Rheumatoid Arthritis and Interstitial Lung Disease: A Prospective Cohort Study
- Author
-
Natalia Mena-Vázquez, María Isabel Padin-Martín, Antonio Fernández-Nebro, C.M. Romero-Barco, Rafaela Ortega-Castro, Blanca Panero Lamothe, Rocio Redondo-Rodriguez, Francisco Javier Godoy-Navarrete, María Carmen Aguilar-Hurtado, Espildora Francisco, F. G. Jiménez-Núñez, M. Rojas-Giménez, Inmaculada Ureña-Garnica, Isabel Añón-Oñate, Sara Manrique-Arija, Lorena Pérez-Albaladejo, María Luisa Velloso-Feijoó, [Mena-Vázquez,N, Romero-Barco,CM, Manrique-Arija,S, Godoy-Navarrete,FJ, Ureña-Garnica,I, Redondo-Rodriguez,R, Jimenez-Núñez,FG, Fernández-Nebro,A] Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain. [Mena-Vázquez,N, Fernández-Nebro,A] UGC de Reumatología, Hospital Regional Universitario de Málaga, Málaga, Spain. [Rojas-Gimenez,M, Ortega-Castro,R] Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain. [Rojas-Gimenez,M, Ortega-Castro,R] UGC de Reumatología, Hospital Universitario Reina Sofía de Córdoba, Córdoba, Spain. [Romero-Barco,CM, Panero Lamothe,B] UGC de Reumatología, Hospital Clínico Universitario Virgen de la Victoria, Málaga, Spain. [Francisco,E] UGC de Neumología, Hospital Regional Universitario de Málaga, Málaga, Spain. [Aguilar-Hurtado,MC, Padin-Martín,MI] UGC de Radiodiagnóstico, Hospital Regional Universitario de Málaga, Málaga, Spain. [Añón-Oñate,I] Hospital Universitario de Jaén, Jaén, Spain. [Pérez-Albaladejo,L] Hospital Universitario Virgen de las Nieves, Granada, Spain. [Velloso-Feijoo,ML] Hospital Universitario Virgen de Valme, Sevilla, Spain. [Fernández-Nebro,A] Departamento de Medicina, Universidad de Málaga, Málaga, Spain, and Grant for Medical Researchers of the 'Fundación Española de Reumatología' 2019.
- Subjects
rheumatoid arthritis ,Tomografía ,Vital capacity ,Antirheumatic drug ,Psychiatry and Psychology::Behavior and Behavior Mechanisms::Behavior::Tobacco Use::Smoking [Medical Subject Headings] ,lcsh:Medicine ,Non-anti-TNF biologics ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Statistics as Topic::Probability::Risk::Risk Factors [Medical Subject Headings] ,Diseases::Respiratory Tract Diseases::Lung Diseases::Lung Diseases, Interstitial [Medical Subject Headings] ,Pulmonary function testing ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,0302 clinical medicine ,Information Science::Information Science::Data Collection::Vital Statistics::Morbidity::Incidence [Medical Subject Headings] ,Usual interstitial pneumonia ,DLCO ,Diffusing capacity ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Diagnostic Techniques, Respiratory System::Respiratory Function Tests [Medical Subject Headings] ,Medicine ,Fumar ,030212 general & internal medicine ,Prospective cohort study ,Tomography ,interstitial lung disease ,non–anti-TNF biologics ,Smoking ,Interstitial lung disease ,General Medicine ,respiratory system ,Diseases::Musculoskeletal Diseases::Rheumatic Diseases::Arthritis, Rheumatoid [Medical Subject Headings] ,Antirreumáticos ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Diagnostic Imaging::Tomography [Medical Subject Headings] ,medicine.medical_specialty ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Epidemiologic Study Characteristics as Topic::Epidemiologic Studies::Cohort Studies::Longitudinal Studies::Prospective Studies [Medical Subject Headings] ,Estudios de cohortes ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Statistics as Topic::Analysis of Variance::Multivariate Analysis [Medical Subject Headings] ,Biologics ,Artritis reumatoide ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Diagnosis::Diagnostic Techniques and Procedures::Diagnostic Techniques, Respiratory System::Respiratory Function Tests::Lung Volume Measurements::Total Lung Capacity::Vital Capacity [Medical Subject Headings] ,Article ,Chemicals and Drugs::Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antirheumatic Agents [Medical Subject Headings] ,03 medical and health sciences ,FEV1/FVC ratio ,Enfermedades pulmonares intersticiales ,Internal medicine ,Productos biológicos ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Epidemiologic Study Characteristics as Topic::Epidemiologic Studies::Cohort Studies::Longitudinal Studies::Follow-Up Studies [Medical Subject Headings] ,biologics ,Rheumatoid arthritis ,Diseases::Respiratory Tract Diseases::Lung Diseases::Lung Diseases, Interstitial::Idiopathic Interstitial Pneumonias::Idiopathic Pulmonary Fibrosis [Medical Subject Headings] ,030203 arthritis & rheumatology ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Statistics as Topic::Regression Analysis [Medical Subject Headings] ,Diseases::Pathological Conditions, Signs and Symptoms::Pathologic Processes::Disease Attributes::Disease Progression [Medical Subject Headings] ,business.industry ,non-anti-TNF biologics ,lcsh:R ,Anatomy::Respiratory System::Lung [Medical Subject Headings] ,medicine.disease ,respiratory tract diseases ,Capacidad vital ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Epidemiologic Methods::Epidemiologic Study Characteristics as Topic::Epidemiologic Studies::Cohort Studies [Medical Subject Headings] ,Chemicals and Drugs::Inorganic Chemicals::Carbon Compounds, Inorganic::Carbon Monoxide [Medical Subject Headings] ,business - Abstract
Objectives: To describe a prospective cohort of patients with rheumatoid arthritis associated with interstitial lung disease (RA-ILD) and identify risk factors associated with disease progression and mortality in this cohort. Patients and methods: We performed a multicenter, prospective, observational study of patients with RA-ILD receiving disease-modifying antirheumatic drugs (DMARDs) between 2015 and 2020. The patients were assessed using high-resolution computed tomography and pulmonary function tests at baseline and at 60 months. The main endpoint was “Progression to ILD at the end of follow-up” in terms of the following outcomes: (1) improvement (i.e., improvement in forced vital capacity (FVC) ≥ 10% or diffusing capacity of the lungs for carbon monoxide (DLCO) ≥ 15% and absence of radiological progression), (2) nonprogression (stabilization or improvement in FVC ≤ 10% or diffusing capacity of the lungs for carbon monoxide (DLCO) <, 15% and absence of radiological progression), (3) progression (worsening of FVC >, 10% or DLCO >, 15% and radiological progression), or (4) death. We recorded demographic and clinical characteristics, lung function, and the incidence of adverse events. A Cox regression analysis was performed to identify factors associated with the worsening of ILD. Results: After 60 months, lung disease had stabilized in 66 patients (56.9%), improved in 9 (7.8%), and worsened in 23 (19.8%). Eighteen patients (15.5%) died, with a mean survival of 71.8 (1.9) months after diagnosis of ILD. The Cox multivariate analysis revealed the independent predictors of worsening of RA-ILD to be usual interstitial pneumonia (hazard ratio (HR), 2.6 (95%CI, 1.0–6.7)), FVC <, 80% (HR, 3.8 (95%CI, 1.5–6.7)), anticitrullinated protein antibody titers (HR, 2.8 (95%CI, 1.1–6.8)), smoking (HR, 2.5 (95%CI, 1.1–6.2)), and treatment with abatacept, tocilizumab, or rituximab (HR, 0.4 (95%CI, 0.2–0.8)). During follow-up, 79 patients (68%) experienced an adverse event, mostly infection (61%). Infection was fatal in 10/18 patients (55.5%) during follow-up. Conclusions: Lung function is stable in most patients with RA-ILD receiving treatment with disease-modifying anti-rheumatic drugs (DMARDs), although one-third worsened or died. Identifying factors associated with worsening in RA-ILD is important for clinical management.
- Published
- 2021
24. [End-of-life, palliative care and dialysis: ensuring shared decision-making].
- Author
-
Atinault A, Baudelot C, Blot F, Caillé Y, Cléro B, Déjean M, Genon C, Lamothe B, Mercier S, Payen S, Percio S, Quignette N, and Tenaillon A
- Subjects
- Humans, Renal Dialysis, Palliative Care, Death, Decision Making, Terminal Care, Kidney Failure, Chronic therapy
- Abstract
Competing Interests: Les auteurs déclarent n’avoir aucun lien d’intérêts.
- Published
- 2024
25. Genetic Polymorphisms of GGH and ABCC2 Are Associated with Methotrexate Intolerance in Patients with Rheumatoid Arthritis.
- Author
-
Escudero-Contreras A, López-Medina C, Collantes-Estévez E, Ortega-Castro R, Calvo-Gutiérrez J, Mena-Vázquez N, Panero-Lamothe B, Manzanares-Martín B, Cáliz-Cáliz R, Jiménez-Morales A, Ruiz-Jiménez M, and Font-Ugalde P
- Abstract
Objective: to identify new single-nucleotide polymorphisms (SNPs) in genes encoding proteins involved in methotrexate (MTX) metabolism and to evaluate the associations of these SNPs with MTX toxicity or intolerance in a southern Spanish cohort of patients with rheumatoid arthritis (RA)., Methods: An observational, retrospective, and multicenter study was conducted at three participating hospitals in southern Spain. The main variable was intolerance to MTX (i.e., bDMARD monotherapy), defined as an interruption of treatment due to adverse events or toxicity. Patients being treated with MTX and bDMARDs (combined treatment) at the time of the study visit were considered "tolerant" of MTX. Ten polymorphisms were selected for sequencing in our patients according to a literature review. Each polymorphism was classified according to three possible genotypes (e.g., two homozygous (AA or GG) and one heterozygous (AG)), and the association of these combinations with MTX intolerance was evaluated., Results: A total of 227 patients were included in the final analysis (107 intolerant of MTX and 120 tolerant). A significant association was observed between MTX intolerance and the GGH-T401C AA/AG genotype (OR 2.13, 95% CI 1.06-4.29) in comparison with the GG genotype. On the other hand, an inverse association was observed between the ABCC2-C24T TT/TC genotype and intolerance to MTX (OR 0.59, 95% CI 0.35-1.00) in comparison with the CC genotype., Conclusion: This study provides new data on the association between genetic polymorphisms and MTX intolerance, which may contribute to the development of new biomarkers and personalized medicine in patients with RA.
- Published
- 2021
- Full Text
- View/download PDF
26. Predictors of Progression and Mortality in Patients with Prevalent Rheumatoid Arthritis and Interstitial Lung Disease: A Prospective Cohort Study.
- Author
-
Mena-Vázquez N, Rojas-Gimenez M, Romero-Barco CM, Manrique-Arija S, Francisco E, Aguilar-Hurtado MC, Añón-Oñate I, Pérez-Albaladejo L, Ortega-Castro R, Godoy-Navarrete FJ, Ureña-Garnica I, Velloso-Feijoo ML, Redondo-Rodriguez R, Jimenez-Núñez FG, Panero Lamothe B, Padin-Martín MI, and Fernández-Nebro A
- Abstract
Objectives: To describe a prospective cohort of patients with rheumatoid arthritis associated with interstitial lung disease (RA-ILD) and identify risk factors associated with disease progression and mortality in this cohort., Patients and Methods: We performed a multicenter, prospective, observational study of patients with RA-ILD receiving disease-modifying antirheumatic drugs (DMARDs) between 2015 and 2020. The patients were assessed using high-resolution computed tomography and pulmonary function tests at baseline and at 60 months. The main endpoint was "Progression to ILD at the end of follow-up" in terms of the following outcomes: (1) improvement (i.e., improvement in forced vital capacity (FVC) ≥10% or diffusing capacity of the lungs for carbon monoxide (DLCO) ≥15% and absence of radiological progression); (2) nonprogression (stabilization or improvement in FVC ≤10% or diffusing capacity of the lungs for carbon monoxide (DLCO) <15% and absence of radiological progression); (3) progression (worsening of FVC >10% or DLCO >15% and radiological progression); or (4) death. We recorded demographic and clinical characteristics, lung function, and the incidence of adverse events. A Cox regression analysis was performed to identify factors associated with the worsening of ILD., Results: After 60 months, lung disease had stabilized in 66 patients (56.9%), improved in 9 (7.8%), and worsened in 23 (19.8%). Eighteen patients (15.5%) died, with a mean survival of 71.8 (1.9) months after diagnosis of ILD. The Cox multivariate analysis revealed the independent predictors of worsening of RA-ILD to be usual interstitial pneumonia (hazard ratio (HR), 2.6 (95%CI, 1.0-6.7)), FVC <80% (HR, 3.8 (95%CI, 1.5-6.7)), anticitrullinated protein antibody titers (HR, 2.8 (95%CI, 1.1-6.8)), smoking (HR, 2.5 (95%CI, 1.1-6.2)), and treatment with abatacept, tocilizumab, or rituximab (HR, 0.4 (95%CI, 0.2-0.8)). During follow-up, 79 patients (68%) experienced an adverse event, mostly infection (61%). Infection was fatal in 10/18 patients (55.5%) during follow-up., Conclusions: Lung function is stable in most patients with RA-ILD receiving treatment with disease-modifying anti-rheumatic drugs (DMARDs), although one-third worsened or died. Identifying factors associated with worsening in RA-ILD is important for clinical management.
- Published
- 2021
- Full Text
- View/download PDF
27. A Pilot Phase II Study of Erlotinib for the Treatment of Patients with Relapsed/Refractory Acute Myeloid Leukemia.
- Author
-
Abou Dalle I, Cortes JE, Pinnamaneni P, Lamothe B, Diaz Duque A, Randhawa J, Pemmaraju N, Jabbour E, Ferrajoli A, Wierda WG, Estrov Z, Konopleva M, Ravandi F, Alvarado Y, Borthakur G, Gandhi V, and Kantarjian HM
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents adverse effects, Biomarkers, Tumor metabolism, Disease-Free Survival, Drug Administration Schedule, Erlotinib Hydrochloride adverse effects, Fatigue etiology, Female, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Recurrence, Treatment Outcome, Young Adult, Antineoplastic Agents therapeutic use, Erlotinib Hydrochloride therapeutic use, Leukemia, Myeloid, Acute drug therapy
- Abstract
Erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, may have off-target activity inducing acute myeloid leukemia (AML) differentiation, possibly through SYK inhibition. We investigated erlotinib in a pilot phase II study for efficacy in relapsed/refractory AML patients at a dose of 150 mg once daily in 28-day cycles. Twenty-nine patients were treated for a median of 29 days (range 12-142 days). Seven patients (24%) received > 1 cycle of therapy and 12 (41%) discontinued treatment before day 28 due to disease progression. One patient (3%) achieved complete remission and 2 (7%) a > 50% reduction in blasts. The most common toxicities associated with erlotinib were fatigue in 10 patients (34%), diarrhea in 10 (34%), nausea in 8 (28%), and rash in 7 (24%). Only 2 patients (7%) had study drug-related adverse events requiring dose reductions and eventual discontinuation. The main reason for treatment discontinuation was disease progression in 26 patients (90%). All patients had died by the time of the last follow-up. Progression of disease was the primary cause of death in all patients. Median overall survival was 14 weeks (range 2.3-96.9 weeks) and median event-free survival was 5 weeks (range 1.7-21.0 weeks). Erlotinib as a single agent has limited clinical efficacy in patients with relapsed/refractory AML., (© 2018 S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
28. Carfilzomib Triggers Cell Death in Chronic Lymphocytic Leukemia by Inducing Proapoptotic and Endoplasmic Reticulum Stress Responses.
- Author
-
Lamothe B, Wierda WG, Keating MJ, and Gandhi V
- Subjects
- Adult, Aged, Ataxia Telangiectasia Mutated Proteins metabolism, Biomarkers, Cell Line, Tumor, Female, Humans, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell therapy, Male, Middle Aged, Mutation, Proteasome Endopeptidase Complex metabolism, Protein Binding, Transcription Factor CHOP metabolism, Ubiquitinated Proteins metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, Endoplasmic Reticulum Stress drug effects, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Oligopeptides pharmacology
- Abstract
Purpose: Carfilzomib, while active in B-cell neoplasms, displayed heterogeneous response in chronic lymphocytic leukemia (CLL) samples from patients and showed interpatient variability to carfilzomib-induced cell death. To understand this variability and predict patients who would respond to carfilzomib, we investigated the mechanism by which carfilzomib induces CLL cell death., Experimental Design: Using CLL patient samples and cell lines, complementary knockdown and knockout cells, and carfilzomib-resistant cell lines, we evaluated changes in intracellular networks to identify molecules responsible for carfilzomib's cytotoxic activity. Lysates from carfilzomib-treated cells were immunoblotted for molecules involved in ubiquitin, apoptotic, and endoplasmic reticulum (ER) stress response pathways and results correlated with carfilzomib cytotoxic activity. Coimmunoprecipitation and pull-down assays were performed to identify complex interactions among MCL-1, Noxa, and Bak., Results: Carfilzomib triggered ER stress and activation of both the intrinsic and extrinsic apoptotic pathways through alteration of the ubiquitin proteasome pathway. Consequently, the transcription factor CCAAT/enhancer-binding protein homology protein (CHOP) accumulated in response to carfilzomib, and CHOP depletion conferred protection against cytotoxicity. Carfilzomib also induced accumulation of MCL-1 and Noxa, whereby MCL-1 preferentially formed a complex with Noxa and consequently relieved MCL-1's protective effect on sequestering Bak. Accordingly, depletion of Noxa or both Bak and Bax conferred protection against carfilzomib-induced cell death., Conclusions: Collectively, carfilzomib induced ER stress culminating in activation of intrinsic and extrinsic caspase pathways, and we identified the CHOP protein level as a biomarker that could predict sensitivity to carfilzomib in CLL. Clin Cancer Res; 22(18); 4712-26. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
29. Pharmacological and Protein Profiling Suggests Venetoclax (ABT-199) as Optimal Partner with Ibrutinib in Chronic Lymphocytic Leukemia.
- Author
-
Cervantes-Gomez F, Lamothe B, Woyach JA, Wierda WG, Keating MJ, Balakrishnan K, and Gandhi V
- Subjects
- Adenine analogs & derivatives, Agammaglobulinaemia Tyrosine Kinase, Aged, Apoptosis drug effects, B-Cell Activating Factor biosynthesis, B-Cell Activating Factor genetics, Bendamustine Hydrochloride administration & dosage, Biphenyl Compounds administration & dosage, Female, Gene Expression Regulation, Leukemic drug effects, Humans, Isoquinolines administration & dosage, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Male, Neoplastic Cells, Circulating drug effects, Neoplastic Cells, Circulating metabolism, Nitrophenols administration & dosage, Piperazines administration & dosage, Piperidines, Protein-Tyrosine Kinases biosynthesis, Protein-Tyrosine Kinases genetics, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Purines administration & dosage, bcl-X Protein biosynthesis, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Proto-Oncogene Proteins c-bcl-2 genetics, Pyrazoles administration & dosage, Pyrimidines administration & dosage, Sulfonamides administration & dosage
- Abstract
Purpose: Bruton's tyrosine kinase (BTK) is a critical enzyme in the B-cell receptor pathway and is inhibited by ibrutinib due to covalent binding to the kinase domain. Though ibrutinib results in impressive clinical activity in chronic lymphocytic leukemia (CLL), most patients achieve only partial remission due to residual disease. We performed a pharmacologic profiling of residual circulating CLL cells from patients receiving ibrutinib to identify optimal agents that could induce cell death of these lymphocytes., Experimental Design: Ex vivo serial samples of CLL cells from patients on ibrutinib were obtained prior and after (weeks 2, 4, and 12) the start of treatment. These cells were incubated with PI3K inhibitors (idelalisib or IPI-145), bendamustine, additional ibrutinib, or BCL-2 antagonists (ABT-737 or ABT-199), and cell death was measured. In vitro investigations complemented ex vivo studies. Immunoblots for BTK signaling pathway and antiapoptotic proteins were performed., Results: The BCL-2 antagonists, especially ABT-199, induced high cell death during ex vivo incubations. In concert with the ex vivo data, in vitro combinations also resulted in high cytotoxicity. Serial samples of CLL cells obtained before and 2, 4, 12, or 36 weeks after the start of ibrutinib showed inhibition of BTK activity and sensitivity to ABTs. Among the three BCL-2 family antiapoptotic proteins that are overexpressed in CLL, levels of MCL-1 and BCL-XL were decreased after ibrutinib while ABT-199 selectively antagonizes BCL-2., Conclusions: Our biologic and molecular results suggest that ibrutinib and ABT-199 combination should be tested clinically against CLL., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
30. Proteasome inhibitor carfilzomib complements ibrutinib's action in chronic lymphocytic leukemia.
- Author
-
Lamothe B, Cervantes-Gomez F, Sivina M, Wierda WG, Keating MJ, and Gandhi V
- Subjects
- Adenine analogs & derivatives, Aged, Apoptosis drug effects, Blotting, Western, Female, Humans, In Vitro Techniques, Male, Middle Aged, Oligopeptides administration & dosage, Piperidines, Proteasome Inhibitors administration & dosage, Pyrazoles administration & dosage, Pyrimidines administration & dosage, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Leukemia, Lymphocytic, Chronic, B-Cell
- Published
- 2015
- Full Text
- View/download PDF
31. Reducing the need for central dual-energy X-ray absorptiometry in postmenopausal women: efficacy of a clinical algorithm including peripheral densitometry.
- Author
-
Jiménez-Núñez FG, Manrique-Arija S, Ureña-Garnica I, Romero-Barco CM, Panero-Lamothe B, Descalzo MA, Carmona L, Rodríguez-Pérez M, and Fernández-Nebro A
- Subjects
- Absorptiometry, Photon, Bone Density, Cross-Sectional Studies, Densitometry, Female, Humans, Middle Aged, Osteoporosis, Postmenopausal epidemiology, Risk Assessment, Algorithms, Osteoporosis, Postmenopausal diagnostic imaging
- Abstract
We evaluated the efficacy of a triage approach based on a combination of osteoporosis risk-assessment tools plus peripheral densitometry to identify low bone density accurately enough to be useful for clinical decision making in postmenopausal women. We conducted a cross-sectional diagnostic study in postmenopausal Caucasian women from primary and tertiary care. All women underwent dual-energy X-ray absorptiometric (DXA) measurement at the hip and lumbar spine and were categorized as osteoporotic or not. Additionally, patients had a nondominant heel densitometry performed with a PIXI densitometer. Four osteoporosis risk scores were tested: SCORE, ORAI, OST, and OSIRIS. All measurements were cross-blinded. We estimated the area under the curve (AUC) to predict the DXA results of 16 combinations of PIXI plus risk scores. A formula including the best combination was derived from a regression model and its predictability estimated. We included 505 women, in whom the prevalence of osteoporosis was 20 %, similar in both settings. The best algorithm was a combination of PIXI + OST + SCORE with an AUC of 0.826 (95 % CI 0.782-0.869). The proposed formula is Risk = (-12) × [PIXI + (-5)] × [OST + (-2)] × SCORE and showed little bias in the estimation (0.0016). If the formula had been implemented and the intermediate risk cutoff set at -5 to 20, the system would have saved
4,606.34 in the study year. The formula proposed, derived from previously validated risk scores plus a peripheral bone density measurement, can be used reliably in primary care to avoid unnecessary central DXA measurements in postmenopausal women. - Published
- 2013
- Full Text
- View/download PDF
32. TAK1 is essential for osteoclast differentiation and is an important modulator of cell death by apoptosis and necroptosis.
- Author
-
Lamothe B, Lai Y, Xie M, Schneider MD, and Darnay BG
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Cell Differentiation, Cell Line, Cell Survival, Cells, Cultured, Gene Deletion, Gene Knockout Techniques, MAP Kinase Kinase Kinases genetics, Mice, Monocytes metabolism, Osteoclasts metabolism, Osteogenesis, RANK Ligand metabolism, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Receptors, Tumor Necrosis Factor, Type I genetics, Tumor Necrosis Factor-alpha metabolism, Apoptosis, MAP Kinase Kinase Kinases metabolism, Monocytes cytology, Necrosis, Osteoclasts cytology
- Abstract
Transforming growth factor β (TGF-β)-activated kinase 1 (TAK1), a mitogen-activated protein 3 (MAP3) kinase, plays an essential role in inflammation by activating the IκB kinase (IKK)/nuclear factor κB (NF-κB) and stress kinase (p38 and c-Jun N-terminal kinase [JNK]) pathways in response to many stimuli. The tumor necrosis factor (TNF) superfamily member receptor activator of NF-κB ligand (RANKL) regulates osteoclastogenesis through its receptor, RANK, and the signaling adaptor TRAF6. Because TAK1 activation is mediated through TRAF6 in the interleukin 1 receptor (IL-1R) and toll-like receptor (TLR) pathways, we sought to investigate the consequence of TAK1 deletion in RANKL-mediated osteoclastogenesis. We generated macrophage colony-stimulating factor (M-CSF)-derived monocytes from the bone marrow of mice with TAK1 deletion in the myeloid lineage. Unexpectedly, TAK1-deficient monocytes in culture died rapidly but could be rescued by retroviral expression of TAK1, inhibition of receptor-interacting protein 1 (RIP1) kinase activity with necrostatin-1, or simultaneous genetic deletion of TNF receptor 1 (TNFR1). Further investigation using TAK1-deficient mouse embryonic fibroblasts revealed that TNF-α-induced cell death was abrogated by the simultaneous inhibition of caspases and knockdown of RIP3, suggesting that TAK1 is an important modulator of both apoptosis and necroptosis. Moreover, TAK1-deficient monocytes rescued from programmed cell death did not form mature osteoclasts in response to RANKL, indicating that TAK1 is indispensable to RANKL-induced osteoclastogenesis. To our knowledge, we are the first to report that mice in which TAK1 has been conditionally deleted in osteoclasts develop osteopetrosis.
- Published
- 2013
- Full Text
- View/download PDF
33. Deletion of TAK1 in the myeloid lineage results in the spontaneous development of myelomonocytic leukemia in mice.
- Author
-
Lamothe B, Lai Y, Hur L, Orozco NM, Wang J, Campos AD, Xie M, Schneider MD, Lockworth CR, Jakacky J, Tran D, Ho M, Dawud S, Dong C, Lin HK, Hu P, Estrov Z, Bueso-Ramos CE, and Darnay BG
- Subjects
- Animals, Cytokines physiology, Flow Cytometry, In Situ Hybridization, Fluorescence, Karyotyping, Mice, Mice, Knockout, Signal Transduction, Splenomegaly genetics, Gene Deletion, Leukemia, Myelomonocytic, Acute genetics, MAP Kinase Kinase Kinases genetics
- Abstract
Previous studies of the conditional ablation of TGF-β activated kinase 1 (TAK1) in mice indicate that TAK1 has an obligatory role in the survival and/or development of hematopoietic stem cells, B cells, T cells, hepatocytes, intestinal epithelial cells, keratinocytes, and various tissues, primarily because of these cells' increased apoptotic sensitivity, and have implicated TAK1 as a critical regulator of the NF-κB and stress kinase pathways and thus a key intermediary in cellular survival. Contrary to this understanding of TAK1's role, we report a mouse model in which TAK1 deletion in the myeloid compartment that evoked a clonal myelomonocytic cell expansion, splenomegaly, multi-organ infiltration, genomic instability, and aggressive, fatal myelomonocytic leukemia. Unlike in previous reports, simultaneous deletion of TNF receptor 1 (TNFR1) failed to rescue this severe phenotype. We found that the features of the disease in our mouse model resemble those of human chronic myelomonocytic leukemia (CMML) in its transformation to acute myeloid leukemia (AML). Consequently, we found TAK1 deletion in 13 of 30 AML patients (43%), thus providing direct genetic evidence of TAK1's role in leukemogenesis.
- Published
- 2012
- Full Text
- View/download PDF
34. Virion assembly factories in the nucleus of polyomavirus-infected cells.
- Author
-
Erickson KD, Bouchet-Marquis C, Heiser K, Szomolanyi-Tsuda E, Mishra R, Lamothe B, Hoenger A, and Garcea RL
- Subjects
- 3T3 Cells, Animals, Cell Nucleus genetics, Cell Nucleus metabolism, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, DNA, Viral genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Embryo, Mammalian metabolism, Embryo, Mammalian pathology, Embryo, Mammalian virology, Fibroblasts metabolism, Fibroblasts pathology, Fibroblasts virology, MRE11 Homologue Protein, Mice, Mice, Knockout, Nuclear Proteins genetics, Polyomavirus Infections genetics, Promyelocytic Leukemia Protein, Transcription Factors genetics, Tumor Suppressor Proteins genetics, Cell Nucleus virology, DNA, Viral metabolism, Nuclear Proteins metabolism, Polyomavirus physiology, Polyomavirus Infections metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism, Virus Assembly physiology
- Abstract
Most DNA viruses replicate in the cell nucleus, although the specific sites of virion assembly are as yet poorly defined. Electron microscopy on freeze-substituted, plastic-embedded sections of murine polyomavirus (PyV)-infected 3T3 mouse fibroblasts or mouse embryonic fibroblasts (MEFs) revealed tubular structures in the nucleus adjacent to clusters of assembled virions, with virions apparently "shed" or "budding" from their ends. Promyelocytic leukemia nuclear bodies (PML-NBs) have been suggested as possible sites for viral replication of polyomaviruses (BKV and SV40), herpes simplex virus (HSV), and adenovirus (Ad). Immunohistochemistry and FISH demonstrated co-localization of the viral T-antigen (Tag), PyV DNA, and the host DNA repair protein MRE11, adjacent to the PML-NBs. In PML⁻/⁻ MEFs the co-localization of MRE11, Tag, and PyV DNA remained unchanged, suggesting that the PML protein itself was not responsible for their association. Furthermore, PyV-infected PML⁻/⁻ MEFs and PML⁻/⁻ mice replicated wild-type levels of infectious virus. Therefore, although the PML protein may identify sites of PyV replication, neither the observed "virus factories" nor virus assembly were dependent on PML. The ultrastructure of the tubes suggests a new model for the encapsidation of small DNA viruses.
- Published
- 2012
- Full Text
- View/download PDF
35. Critical role of monoubiquitination of histone H2AX protein in histone H2AX phosphorylation and DNA damage response.
- Author
-
Wu CY, Kang HY, Yang WL, Wu J, Jeong YS, Wang J, Chan CH, Lee SW, Zhang X, Lamothe B, Campos AD, Darnay BG, and Lin HK
- Subjects
- Animals, Binding Sites, Cell Line, Tumor, DNA Repair, Histones physiology, Humans, Mice, Models, Biological, Phosphorylation, Radiation, Ionizing, Signal Transduction, Transfection, Ubiquitin metabolism, DNA Damage, Histones metabolism, Neoplasms metabolism, Ubiquitin chemistry
- Abstract
DNA damage response is an important surveillance mechanism used to maintain the integrity of the human genome in response to genotoxic stress. Histone variant H2AX is a critical sensor that undergoes phosphorylation at serine 139 upon genotoxic stress, which provides a docking site to recruit the mediator of DNA damage checkpoint protein 1 (MDC1) and DNA repair protein complex to sites of DNA breaks for DNA repair. Here, we show that monoubiquitination of H2AX is induced upon DNA double strand breaks and plays a critical role in H2AX Ser-139 phosphorylation (γ-H2AX), in turn facilitating the recruitment of MDC1 to DNA damage foci. Mechanistically, we show that monoubiquitination of H2AX induced by RING finger protein 2 (RNF2) is required for the recruitment of active ataxia telangiectasia mutated to DNA damage foci, thus affecting the formation of γ-H2AX. Importantly, a defect in monoubiquitination of H2AX profoundly enhances ionizing radiation sensitivity. Our study therefore suggests that monoubiquitination of H2AX is an important step for DNA damage response and may have important clinical implications for the treatment of cancers.
- Published
- 2011
- Full Text
- View/download PDF
36. Structural basis for the lack of E2 interaction in the RING domain of TRAF2.
- Author
-
Yin Q, Lamothe B, Darnay BG, and Wu H
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Humans, Molecular Sequence Data, Protein Conformation, Sequence Homology, Amino Acid, TNF Receptor-Associated Factor 2 metabolism, Ubiquitination, TNF Receptor-Associated Factor 2 chemistry
- Abstract
TRAF proteins are intracellular signal transducers for a number of immune receptor superfamilies. Specifically, TRAF2 interacts with members of the TNF receptor superfamily and connects the receptors to downstream signaling proteins. It has been assumed that TRAF2 is a ubiquitin ligase like TRAF6 and mediates K63-linked polyubiquitination of RIP1, a kinase pivotal in TNFalpha-induced NF-kappaB activation. Here we report the crystal structure of the RING and the first zinc finger domains of TRAF2. We show that the TRAF2 RING structure is very different from the known TRAF6 RING structure. The differences are multifaceted, including amino acid differences at the critical Ubc13-interacting site, local conformational differences, and a unique nine-residue insertion between the RING domain and the first zinc finger in TRAF2. These structural differences prevent TRAF2 from interacting with Ubc13 and other related E2s via steric clash and unfavorable interfaces. Our structural observation should prompt a re-evaluation of the role of TRAF2 in TNFalpha signaling and may indicate that TRAF2-associated proteins such as cIAPs may be the ubiquitin ligases for NF-kappaB signaling.
- Published
- 2009
- Full Text
- View/download PDF
37. The E3 ligase TRAF6 regulates Akt ubiquitination and activation.
- Author
-
Yang WL, Wang J, Chan CH, Lee SW, Campos AD, Lamothe B, Hur L, Grabiner BC, Lin X, Darnay BG, and Lin HK
- Subjects
- Amino Acid Motifs, Animals, Apoptosis, Cell Line, Cell Line, Tumor, Humans, Insulin-Like Growth Factor I pharmacology, Interleukin-1beta pharmacology, Lipopolysaccharides pharmacology, Mice, Neoplasm Transplantation, Neoplasms, Experimental metabolism, Phosphatidylinositol Phosphates metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt chemistry, TNF Receptor-Associated Factor 6 genetics, Transplantation, Heterologous, Ubiquitination, Cell Membrane metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Akt signaling plays a central role in many biological functions, such as cell proliferation and apoptosis. Because Akt (also known as protein kinase B) resides primarily in the cytosol, it is not known how these signaling molecules are recruited to the plasma membrane and subsequently activated by growth factor stimuli. We found that the protein kinase Akt undergoes lysine-63 chain ubiquitination, which is important for Akt membrane localization and phosphorylation. TRAF6 was found to be a direct E3 ligase for Akt and was essential for Akt ubiquitination, membrane recruitment, and phosphorylation upon growth-factor stimulation. The human cancer-associated Akt mutant displayed an increase in Akt ubiquitination, in turn contributing to the enhancement of Akt membrane localization and phosphorylation. Thus, Akt ubiquitination is an important step for oncogenic Akt activation.
- Published
- 2009
- Full Text
- View/download PDF
38. E2 interaction and dimerization in the crystal structure of TRAF6.
- Author
-
Yin Q, Lin SC, Lamothe B, Lu M, Lo YC, Hura G, Zheng L, Rich RL, Campos AD, Myszka DG, Lenardo MJ, Darnay BG, and Wu H
- Subjects
- Crystallography, X-Ray, Fluorescence Resonance Energy Transfer, Humans, Protein Conformation, Protein Multimerization, RING Finger Domains, Ubiquitination, Zinc Fingers, TNF Receptor-Associated Factor 6 chemistry, Ubiquitin-Conjugating Enzymes chemistry
- Abstract
Tumor necrosis factor (TNF) receptor-associated factor (TRAF)-6 mediates Lys63-linked polyubiquitination for NF-kappaB activation via its N-terminal RING and zinc finger domains. Here we report the crystal structures of TRAF6 and its complex with the ubiquitin-conjugating enzyme (E2) Ubc13. The RING and zinc fingers of TRAF6 assume a rigid, elongated structure. Interaction of TRAF6 with Ubc13 involves direct contacts of the RING and the preceding residues, and the first zinc finger has a structural role. Unexpectedly, this region of TRAF6 is dimeric both in the crystal and in solution, different from the trimeric C-terminal TRAF domain. Structure-based mutagenesis reveals that TRAF6 dimerization is crucial for polyubiquitin synthesis and autoubiquitination. Fluorescence resonance energy transfer analysis shows that TRAF6 dimerization induces higher-order oligomerization of full-length TRAF6. The mismatch of dimeric and trimeric symmetry may provide a mode of infinite oligomerization that facilitates ligand-dependent signal transduction of many immune receptors.
- Published
- 2009
- Full Text
- View/download PDF
39. The RING domain and first zinc finger of TRAF6 coordinate signaling by interleukin-1, lipopolysaccharide, and RANKL.
- Author
-
Lamothe B, Campos AD, Webster WK, Gopinathan A, Hur L, and Darnay BG
- Subjects
- Animals, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Line, Humans, I-kappa B Kinase genetics, I-kappa B Kinase metabolism, Immunity, Innate physiology, Interleukin-1 genetics, Interleukin-1 metabolism, Interleukin-6 genetics, Interleukin-6 metabolism, MAP Kinase Kinase 4 genetics, MAP Kinase Kinase 4 metabolism, Mice, Mice, Knockout, Monocytes cytology, Monocytes metabolism, Osteoclasts cytology, Osteoclasts metabolism, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism, Protein Structure, Tertiary physiology, RANK Ligand genetics, Signal Transduction drug effects, TNF Receptor-Associated Factor 6 genetics, Transcription Factors genetics, Transcription Factors metabolism, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitination drug effects, Ubiquitination physiology, Zinc Fingers physiology, p38 Mitogen-Activated Protein Kinases genetics, p38 Mitogen-Activated Protein Kinases metabolism, Interleukin-1 pharmacology, Lipopolysaccharides pharmacology, RANK Ligand metabolism, Signal Transduction physiology, TNF Receptor-Associated Factor 6 metabolism
- Abstract
TRAF6, a crucial adaptor molecule in innate and adaptive immunity, contains three distinct functional domains. The C-terminal TRAF domain facilitates oligomerization and sequence-specific interaction with receptors or other adaptor proteins. In conjunction with the dimeric E2 enzyme Ubc13-Uev1A, the N-terminal RING domain of TRAF6 functions as an E3 ubiquitin (Ub) ligase that facilitates its own site-specific ubiquitination through the generation of a Lys-63-linked poly-Ub chain. This modification does not cause its proteasomal degradation but rather serves as a scaffold to activate both the IKK and stress kinase pathways. Connecting the N-and C-terminal regions, the four internal zinc finger (ZF) motifs have yet to be functionally defined. In this study, we examined the role of the ZF domains in interleukin-1, lipopolysaccharide, and RANKL signaling by reconstitution of TRAF6-deficient cells with point mutations or deletions of these ZF motifs. Although ZF domains 2-4 are dispensable for activating IKK, p38, and JNK by interleukin-1 and lipopolysaccharide, the first ZF domain together with an intact RING domain of TRAF6 is essential for activating these pathways. Furthermore, TRAF6 autoubiquitination and its interaction with Ubc13 are dependent on ZF1 and an intact RING domain. Additionally, expression of TRAF6 lacking ZF2-4 in TRAF6-deficient monocytes rescues RANKL-mediated osteoclast differentiation and LPS-stimulated interleukin-6 production. These data provide evidence for the critical role of the Ub ligase activity of TRAF6, which is coordinated via the RING domain and ZF1 to supply the necessary elements in signaling by cytokines dependent upon TRAF6.
- Published
- 2008
- Full Text
- View/download PDF
40. Molecular basis for the unique deubiquitinating activity of the NF-kappaB inhibitor A20.
- Author
-
Lin SC, Chung JY, Lamothe B, Rajashankar K, Lu M, Lo YC, Lam AY, Darnay BG, and Wu H
- Subjects
- Alanine metabolism, Amino Acid Sequence, Amino Acid Substitution, Binding Sites, Catalysis, Cell Line, Conserved Sequence, Crystallography, X-Ray, DNA-Binding Proteins, Escherichia coli genetics, Gene Deletion, Glutathione Transferase metabolism, Humans, Hydrogen Bonding, Intracellular Signaling Peptides and Proteins chemistry, Intracellular Signaling Peptides and Proteins genetics, Intracellular Signaling Peptides and Proteins isolation & purification, Kidney cytology, Models, Molecular, Molecular Sequence Data, Nuclear Proteins chemistry, Nuclear Proteins genetics, Nuclear Proteins isolation & purification, Polyubiquitin chemistry, Polyubiquitin metabolism, Precipitin Tests, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Sequence Homology, Amino Acid, Static Electricity, Substrate Specificity, TNF Receptor-Associated Factor 6 metabolism, Tumor Necrosis Factor alpha-Induced Protein 3, Ubiquitin metabolism, Ubiquitin-Protein Ligases chemistry, Intracellular Signaling Peptides and Proteins metabolism, NF-kappa B antagonists & inhibitors, Nuclear Proteins metabolism, Ubiquitination
- Abstract
Nuclear factor kappaB (NF-kappaB) activation in tumor necrosis factor, interleukin-1, and Toll-like receptor pathways requires Lys63-linked nondegradative polyubiquitination. A20 is a specific feedback inhibitor of NF-kappaB activation in these pathways that possesses dual ubiquitin-editing functions. While the N-terminal domain of A20 is a deubiquitinating enzyme (DUB) for Lys63-linked polyubiquitinated signaling mediators such as TRAF6 and RIP, its C-terminal domain is a ubiquitin ligase (E3) for Lys48-linked degradative polyubiquitination of the same substrates. To elucidate the molecular basis for the DUB activity of A20, we determined its crystal structure and performed a series of biochemical and cell biological studies. The structure reveals the potential catalytic mechanism of A20, which may be significantly different from papain-like cysteine proteases. Ubiquitin can be docked onto a conserved A20 surface; this interaction exhibits charge complementarity and no steric clash. Surprisingly, A20 does not have specificity for Lys63-linked polyubiquitin chains. Instead, it effectively removes Lys63-linked polyubiquitin chains from TRAF6 without dissembling the chains themselves. Our studies suggest that A20 does not act as a general DUB but has the specificity for particular polyubiquitinated substrates to assure its fidelity in regulating NF-kappaB activation in the tumor necrosis factor, interleukin-1, and Toll-like receptor pathways.
- Published
- 2008
- Full Text
- View/download PDF
41. TRAF6 ubiquitin ligase is essential for RANKL signaling and osteoclast differentiation.
- Author
-
Lamothe B, Webster WK, Gopinathan A, Besse A, Campos AD, and Darnay BG
- Subjects
- Animals, Cell Differentiation physiology, Cell Line, Humans, Mice, Signal Transduction physiology, Osteoblasts cytology, Osteoblasts metabolism, Osteoclasts cytology, Osteoclasts metabolism, RANK Ligand metabolism, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin-Protein Ligase Complexes metabolism
- Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6), the crucial adaptor molecule of receptor activator of NF-kappaB (RANK), plays an essential role in governing the formation of multi-nucleated osteoclasts. TRAF6 is a RING-dependent ubiquitin (Ub) ligase that in conjunction with Ubc13/Uev1A catalyzes its own auto-ubiquitination via Lys63-linked poly-Ub chains. While the receptor-adaptor function of TRAF6 in RANK signaling is well understood, the significance of its Ub ligase activity in this process remains largely unknown. In this study, we show that retroviral expression of TRAF6, but not a RING mutant of TRAF6 was able to rescue TRAF6-deficient monocytes for the activation of IKK and osteoclast differentiation by RANKL. Furthermore, a catalytically inactive Ubc13 or stable knockdown of Ubc13 significantly prevents RANK-mediated TRAF6 ubiquitination and NF-kappaB and JNK activation. These data establish a signaling cascade in which regulated Lys63-linked TRAF6 auto-ubiquitination is the critical upstream mediator of osteoclast differentiation.
- Published
- 2007
- Full Text
- View/download PDF
42. TAK1-dependent signaling requires functional interaction with TAB2/TAB3.
- Author
-
Besse A, Lamothe B, Campos AD, Webster WK, Maddineni U, Lin SC, Wu H, and Darnay BG
- Subjects
- Adaptor Proteins, Signal Transducing antagonists & inhibitors, Adaptor Proteins, Signal Transducing metabolism, Amino Acid Sequence, Animals, Cell Differentiation genetics, Cell Differentiation physiology, Cell Line, Humans, Interleukin-1 physiology, Intracellular Signaling Peptides and Proteins antagonists & inhibitors, L Cells, MAP Kinase Kinase Kinases genetics, MAP Kinase Kinase Kinases metabolism, Mice, Molecular Sequence Data, Osteoclasts cytology, Osteoclasts physiology, Peptide Fragments genetics, Peptide Fragments metabolism, Peptide Fragments physiology, Protein Binding genetics, Protein Binding physiology, Protein Structure, Tertiary genetics, Protein Structure, Tertiary physiology, RANK Ligand physiology, Sequence Deletion, Signal Transduction genetics, Tumor Necrosis Factor-alpha physiology, Adaptor Proteins, Signal Transducing physiology, Intracellular Signaling Peptides and Proteins physiology, MAP Kinase Kinase Kinases physiology, Signal Transduction physiology
- Abstract
Transforming growth factor beta-activated kinase 1 (TAK1), a member of the MAPKKK family, was initially described to play an essential role in the transforming growth factor beta-signaling pathway, but recent evidence has emerged implicating TAK1 in the interleukin (IL)-1 and tumor necrosis factor (TNF) pathways. Notably, two homologous proteins, TAB2 and TAB3, have been identified as adaptors linking TAK1 to the upstream adaptors TRAFs. However, it remains unclear whether the interaction between TAB2/TAB3 and TAK1 is necessary for its kinase activation and subsequent activation of the IKK and MAPK pathways. Here, we characterized the TAB2/TAB3-binding domain in TAK1 and further examined the requirement of this interaction for IL-1, TNF, and RANKL signaling. Through deletion mapping experiments, we demonstrated that the binding motif for TAB2/TAB3 is a non-contiguous region located within the last C-terminal 100 residues of TAK1. However, residues 479-553 of TAK1 appear to be necessary and sufficient for TAB2/TAB3 interaction. Conversely, residues 574-693 of TAB2 were shown to interact with TAK1. A green fluorescent protein fusion protein containing the last 100 residues of TAK1 (TAK1-C100) abolished the interaction of endogenous TAB2/TAB3 with TAK1, the phosphorylation of TAK1, and prevented the activation of IKK and MAPK induced by IL-1, TNF, and RANKL. Furthermore, TAK1-C100 blocked RANKL-induced nuclear accumulation of NFATc1 and consequently osteoclast differentiation consistent with the ability of a catalytically inactive TAK1 to block RANKL-mediated signaling. Significantly, our study provides evidence that the TAB2/TAB3 interaction with TAK1 is crucial for the activation of signaling cascades mediated by IL-1, TNF, and RANKL.
- Published
- 2007
- Full Text
- View/download PDF
43. Site-specific Lys-63-linked tumor necrosis factor receptor-associated factor 6 auto-ubiquitination is a critical determinant of I kappa B kinase activation.
- Author
-
Lamothe B, Besse A, Campos AD, Webster WK, Wu H, and Darnay BG
- Subjects
- Animals, Catalysis, Cell Line, Enzyme Activation, Humans, Mice, Mice, Knockout, NF-kappa B metabolism, Protein Structure, Tertiary genetics, Signal Transduction physiology, TNF Receptor-Associated Factor 6 deficiency, TNF Receptor-Associated Factor 6 physiology, Transcription Factors metabolism, Transcription Factors physiology, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Conjugating Enzymes physiology, I-kappa B Kinase metabolism, Lysine metabolism, TNF Receptor-Associated Factor 6 chemistry, TNF Receptor-Associated Factor 6 metabolism, Ubiquitin metabolism
- Abstract
Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) is a key mediator in proximal signaling of the interleukin-1/Toll-like receptor and the TNF receptor superfamily. Analysis of TRAF6-deficient mice revealed a fundamental role of TRAF6 in osteoclastogenesis; however, the molecular mechanism underlying TRAF6 signaling in this biological process is not understood. Recent biochemical evidence has indicated that TRAF6 possesses ubiquitin ligase activity that controls the activation of IKK and NF-kappaB. Because these studies are primarily based on cell-free systems, the role of the ubiquitin ligase activity of TRAF6 and its auto-ubiquitination to initiate the NF-kappaB pathway in vivo remain elusive. Here we show that an intact RING domain of TRAF6 in conjunction with the E2 enzyme Ubc13/Uev1A is necessary for Lys-63-linked auto-ubiquitination of TRAF6 and for its ability to activate IKK and NF-kappaB. Furthermore, a RING mutant of TRAF6 abolishes its ability to induce receptor activator of NF-kappaB-independent osteoclast differentiation and nuclear accumulation of the transcription factor NFATc1. Notably, we map the auto-ubiquitination site of TRAF6 to a single Lys residue, which if mutated renders TRAF6 unable to activate transforming growth factor-beta-activated kinase 1 and IKK and to cause spontaneous osteoclast differentiation. Additionally, we provide biochemical and in vivo evidence that TRAF6 serves as an E3 to directly ubiquitinate NEMO. Reconstituting TRAF6-deficent cells with various TRAF6 mutants, we clearly demonstrate the requirement for the TRAF6 RING domain and site-specific auto-ubiquitination of TRAF6 to activate IKK in response to interleukin-1. These data establish a signaling cascade in which regulated site-specific Lys-63-linked TRAF6 auto-ubiquitination is the critical upstream mediator of IKK.
- Published
- 2007
- Full Text
- View/download PDF
44. TRAFs in RANK signaling.
- Author
-
Darnay BG, Besse A, Poblenz AT, Lamothe B, and Jacoby JJ
- Subjects
- Animals, Humans, Receptor Activator of Nuclear Factor-kappa B chemistry, Receptor Activator of Nuclear Factor-kappa B metabolism, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins chemistry, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins metabolism, Receptor Activator of Nuclear Factor-kappa B physiology, Signal Transduction physiology, Tumor Necrosis Factor Receptor-Associated Peptides and Proteins physiology
- Abstract
Members of the tumor necrosis factor (TNF) family govern many diverse physiological and cellular responses including cellular proliferation, differentiation, and apoptosis. Ligands of this family interact through a distinct set of specific receptors that lack enzymatic activity and therefore are dependent on the association of adaptor molecules. One receptor/ligand pair known as receptor activator of nuclear factor-kappa B (RANK) and RANK ligand (RANKL) regulates bone remodeling, mammary gland development, and lymph node organogenesis. RANK interacts with five members of the TNF receptor-associated factor (TRAF) family, of which TRAF6 is indispensable for its signaling capability. An accumulation of evidence from various research laboratories indicates TRAFs, but more importantly TRAF6, is the key to understanding how RANKL links cytoplasmic signaling to the nuclear transcriptional program.
- Published
- 2007
- Full Text
- View/download PDF
45. The docking protein Gab1 is the primary mediator of EGF-stimulated activation of the PI-3K/Akt cell survival pathway.
- Author
-
Mattoon DR, Lamothe B, Lax I, and Schlessinger J
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Cell Survival drug effects, Cells, Cultured, Embryo, Mammalian cytology, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Intracellular Signaling Peptides and Proteins metabolism, Mice, Mice, Knockout, Phosphoproteins drug effects, Phosphoproteins genetics, Protein Tyrosine Phosphatase, Non-Receptor Type 11, Protein Tyrosine Phosphatases metabolism, Epidermal Growth Factor pharmacology, Phosphatidylinositol 3-Kinases metabolism, Phosphoproteins metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction
- Abstract
Background: Gab1 is a docking protein that recruits phosphatidylinositol-3 kinase (PI-3 kinase) and other effector proteins in response to the activation of many receptor tyrosine kinases (RTKs). As the autophosphorylation sites on EGF-receptor (EGFR) do not include canonical PI-3 kinase binding sites, it is thought that EGF stimulation of PI-3 kinase and its downstream effector Akt is mediated by an indirect mechanism., Results: We used fibroblasts isolated from Gab1-/- mouse embryos to explore the mechanism of EGF stimulation of the PI-3 kinase/Akt anti-apoptotic cell signaling pathway. We demonstrate that Gab1 is essential for EGF stimulation of PI-3 kinase and Akt in these cells and that these responses are mediated by complex formation between p85, the regulatory subunit of PI-3 kinase, and three canonical tyrosine phosphorylation sites on Gab1. Furthermore, complex formation between Gab1 and the protein tyrosine phosphatase Shp2 negatively regulates Gab1 mediated PI-3 kinase and Akt activation following EGF-receptor stimulation. We also demonstrate that tyrosine phosphorylation of ErbB3 may lead to recruitment and activation of PI-3 kinase and Akt in Gab1-/- MEFs., Conclusions: The primary mechanism of EGF-induced stimulation of the PI-3 kinase/Akt anti-apoptotic pathway occurs via the docking protein Gab1. However, in cells expressing ErbB3, EGF and neuroregulin can stimulate PI-3 kinase and Akt activation in a Gab1-dependent or Gab1-independent manner.
- Published
- 2004
- Full Text
- View/download PDF
46. The docking protein Gab1 is an essential component of an indirect mechanism for fibroblast growth factor stimulation of the phosphatidylinositol 3-kinase/Akt antiapoptotic pathway.
- Author
-
Lamothe B, Yamada M, Schaeper U, Birchmeier W, Lax I, and Schlessinger J
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Binding Sites, Cell Survival, Cells, Cultured, Fibroblasts, Membrane Proteins genetics, Mice, Mice, Knockout, Mitogen-Activated Protein Kinases metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt, Recombinant Fusion Proteins pharmacology, Fibroblast Growth Factor 1 physiology, Phosphatidylinositol 3-Kinases metabolism, Phosphoproteins physiology, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Signal Transduction
- Abstract
The docking protein Gab1 has been implicated as a mediator of multiple signaling pathways that are activated by a variety of receptor tyrosine kinases and cytokines. We have previously proposed that fibroblast growth factor 1 (FGF1) stimulation of tyrosine phosphorylation of Gab1 and recruitment of phosphatidylinositol (PI) 3-kinase are mediated by an indirect mechanism in which the docking protein fibroblast receptor substrate 2alpha (FRS2alpha) plays a critical role. In this report, we explore the role of Gab1 in FGF1 signaling by using mouse embryo fibroblasts (MEFs) derived from Gab1(-/-) or FRS2alpha(-/-) mice. We demonstrate that Gab1 is essential for FGF1 stimulation of both PI 3-kinase and the antiapoptotic protein kinase Akt, while FGF1-induced mitogen-activated protein kinase (MAPK) stimulation is not affected by Gab1 deficiency. To test the indirect mechanism for FGF1 stimulation of PI 3-kinase and Akt, we use a chimeric docking protein composed of the membrane targeting signal and the phosphotyrosine-binding domain of FRS2alpha fused to the C-terminal portion of Gab1, the region including the binding sites for the complement of signaling proteins that are recruited by Gab1. We demonstrate that expression of the chimeric docking protein in Gab1(-/-) MEFs rescues PI 3-kinase and the Akt responses, while expression of the chimeric docking protein in FRS2alpha(-/-) MEFs rescues stimulation of both Akt and MAPK. These experiments underscore the essential role of Gab1 in FGF1 stimulation of the PI 3-kinase/Akt signaling pathway and provide further support for the indirect mechanism for FGF1 stimulation of PI 3-kinase involving regulated assembly of a multiprotein complex.
- Published
- 2004
- Full Text
- View/download PDF
47. Partial rescue of insulin receptor-deficient mice by transgenic complementation with an activated insulin receptor in the liver.
- Author
-
Baudry A, Jackerott M, Lamothe B, Kozyrev SV, Leroux L, Durel B, Saint-Just S, and Joshi RL
- Subjects
- Animals, Blood Glucose metabolism, Female, Gene Expression Regulation, Genetic Complementation Test, Genotype, Glucose Tolerance Test, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred CBA, Mice, Transgenic, Mutation, Phenylalanine Hydroxylase genetics, Receptor, Insulin metabolism, Recombinant Fusion Proteins genetics, Survival Analysis, Liver metabolism, Receptor, Insulin genetics
- Abstract
Insulin receptor (IR)-deficient mice develop severe diabetes mellitus, diabetic ketoacidosis (DKA) and liver steatosis and die within 1 week after birth. We examined in this work whether the metabolic phenotype of IR(-/-) mutants could be improved by transgenic complementation with IR selectively in the liver. We first generated transgenic mice expressing a human DNA complementary to RNA encoding a truncated constitutively activated form of IR (IRdelta) under the control of liver-specific phenylalanine hydroxylase (PAH) gene promoter. These mice presented more pronounced fasting hypoglycemia and showed slightly improved glucose tolerance as compared to controls. The transgenic mice were crossed with IR(+/-) mutants to generate IR(-/-) mice carrying the PAH-IRDelta transgene. Although such mutants developed glycosuria, DKA was delayed by more than 1 week and survival was prolonged to 8-20 days in approximately 10% of mice. In these partially rescued pups, serum glucose and triglyceride levels were lowered, hepatic glycogen stores were reconstituted and liver steatosis was absent as compared with pups which developed strong DKA and died earlier. Thus, lack of insulin action in the liver is responsible in large part for the metabolic disorders seen in IR(+/-) mice. This study should stimulate interest in therapeutic strategies aimed at improving hepatic function in diabetes.
- Published
- 2002
- Full Text
- View/download PDF
48. The docking protein FRS2alpha controls a MAP kinase-mediated negative feedback mechanism for signaling by FGF receptors.
- Author
-
Lax I, Wong A, Lamothe B, Lee A, Frost A, Hawes J, and Schlessinger J
- Subjects
- Animals, Cell Adhesion, Cell Division, Cell Line, Cell Movement, Electrophoretic Mobility Shift Assay, Fibroblast Growth Factors pharmacology, GRB2 Adaptor Protein, Humans, Membrane Proteins genetics, Mice, Mutation, PC12 Cells, Phosphoproteins genetics, Phosphorylation, Phosphothreonine metabolism, Phosphotyrosine metabolism, Protein Binding drug effects, Proteins metabolism, Rats, Adaptor Proteins, Signal Transducing, Feedback, Physiological, MAP Kinase Signaling System, Membrane Proteins metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, Phosphoproteins metabolism, Receptors, Fibroblast Growth Factor metabolism
- Abstract
The docking protein FRS2alpha functions as a major mediator of signaling by FGF and NGF receptors. Here we demonstrate that, in addition to tyrosine phosphorylation, FRS2alpha is phosphorylated by MAP kinase on multiple threonine residues in response to FGF stimulation or by insulin, EGF, and PDGF, extracellular stimuli that do not induce tyrosine phosphorylation of FRS2alpha. Prevention of FRS2alpha threonine phosphorylation results in constitutive tyrosine phosphorylation of FRS2alpha in unstimulated cells and enhanced tyrosine phosphorylation of FRS2alpha, MAPK stimulation, cell migration, and proliferation in FGF-stimulated cells. Expression of an FRS2alpha mutant deficient in MAPK phosphorylation sites induces anchorage-independent cell growth and colony formation in soft agar. These experiments reveal a novel MAPK-mediated, negative feedback mechanism for control of signaling pathways that are dependent on FRS2 and a mechanism for heterologous control of signaling via FGF receptors.
- Published
- 2002
- Full Text
- View/download PDF
49. Ectopic expression of protein-tyrosine kinase Bcr-Abl suppresses tumor necrosis factor (TNF)-induced NF-kappa B activation and IkappaBalpha phosphorylation. Relationship with down-regulation of TNF receptors.
- Author
-
Mukhopadhyay A, Shishodia S, Suttles J, Brittingham K, Lamothe B, Nimmanapalli R, Bhalla KN, and Aggarwal BB
- Subjects
- Cell Line, Down-Regulation, Humans, JNK Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinases metabolism, NF-KappaB Inhibitor alpha, Phosphorylation, RNA, Messenger analysis, Receptors, Tumor Necrosis Factor genetics, DNA-Binding Proteins metabolism, Fusion Proteins, bcr-abl physiology, I-kappa B Proteins, NF-kappa B metabolism, Receptors, Tumor Necrosis Factor analysis, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Bcr-Abl, the product of the protooncogene bcr-abl, is a constitutively active protein-tyrosine kinase that is highly expressed in chronic myelogenous leukemia and in acute myeloid leukemia cells. Because Bcr-Abl is known to provide mitogenic signals through suppression of apoptosis, we investigated the effect of this oncogene product on signaling by tumor necrosis factor (TNF), a proapoptotic cytokine. We used a bcr-abl-deficient human megakaryocytic leukemia cell line MO7E and an isogenic MBA cell line stably transfected with bcr-abl. Electrophoretic mobility shift assay revealed that TNF activated the nuclear transcription factor NF-kappaB in MO7E cells but not in MBA cells. The impaired NF-kappaB activation in Bcr-Abl-expressing cells was not due to absence of the NF-kappaB proteins p65, p50, or p100 or of IkappaBalpha or IkappaBbeta. Okadaic acid-induced NF-kappaB activation was unaffected by Bcr-Abl expression. TNF induced IkappaBalpha phosphorylation and degradation in MO7E cells but not in MBA cells. The suppression of TNF-induced NF-kappaB activation by Bcr-Abl was not restricted to MBA cells, because ectopic expression of Bcr-Abl in human acute myeloid leukemia HL-60 cells also blocked TNF-induced NF-kappaB activation. When examined for the TNF receptors by the radioreceptor assay, flow cytometry, or Western blot analysis, we found that Bcr-Abl expression down-regulated the expression of the TNF receptors. The RNase protection assay and Northern blot analysis revealed the transcriptional down-regulation of the TNF receptor by Bcr-Abl protein. Overall, these results indicate that ectopic expression of Bcr-Abl interferes with the TNF signaling pathway through the down-regulation of TNF receptors.
- Published
- 2002
- Full Text
- View/download PDF
50. Distinct molecular mechanism for initiating TRAF6 signalling.
- Author
-
Ye H, Arron JR, Lamothe B, Cirilli M, Kobayashi T, Shevde NK, Segal D, Dzivenu OK, Vologodskaia M, Yim M, Du K, Singh S, Pike JW, Darnay BG, Choi Y, and Wu H
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Binding Sites, CD40 Antigens chemistry, CD40 Antigens genetics, CD40 Antigens metabolism, Cell Differentiation, Cell Line, Crystallography, X-Ray, Humans, Interleukin-1 Receptor-Associated Kinases, Mice, Models, Molecular, Monocytes, Mutation, NF-kappa B metabolism, Osteoclasts cytology, Peptide Fragments chemistry, Peptide Fragments metabolism, Protein Binding, Protein Conformation, Protein Kinases metabolism, Protein Structure, Tertiary, Proteins genetics, Receptor Activator of Nuclear Factor-kappa B, Receptors, Tumor Necrosis Factor chemistry, Receptors, Tumor Necrosis Factor genetics, Receptors, Tumor Necrosis Factor metabolism, TNF Receptor-Associated Factor 2, TNF Receptor-Associated Factor 6, Proteins chemistry, Proteins metabolism, Signal Transduction
- Abstract
Tumour-necrosis factor (TNF) receptor-associated factor 6 (TRAF6) is the only TRAF family member that participates in signal transduction of both the TNF receptor (TNFR) superfamily and the interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) superfamily; it is important for adaptive immunity, innate immunity and bone homeostasis. Here we report crystal structures of TRAF6, alone and in complex with TRAF6-binding peptides from CD40 and TRANCE-R (also known as RANK), members of the TNFR superfamily, to gain insight into the mechanism by which TRAF6 mediates several signalling cascades. A 40 degrees difference in the directions of the bound peptides in TRAF6 and TRAF2 shows that there are marked structural differences between receptor recognition by TRAF6 and other TRAFs. The structural determinant of the petide TRAF6 interaction reveals a Pro-X-Glu-X-X-(aromatic/acidic residue) TRAF6-binding motif, which is present not only in CD40 and TRANCE-R but also in the three IRAK adapter kinases for IL-1R/TLR signalling. Cell-permeable peptides with the TRAF6-binding motif inhibit TRAF6 signalling, which indicates their potential as therapeutic modulators. Our studies identify a universal mechanism by which TRAF6 regulates several signalling cascades in adaptive immunity, innate immunity and bone homeostasis.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.