Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 26(5): 1053-70, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25270074

RESUMEN

TNF ligand superfamily member 12, also known as TNF-related weak inducer of apoptosis (TWEAK), acts through its receptor, fibroblast growth factor-inducible 14 (Fn14), to mediate several key pathologic processes involved in tissue injury relating to lupus nephritis. To explore the potential for renal protection in lupus nephritis by targeting this pathway, we introduced the Fn14 null allele into the MRL-lpr/lpr lupus mouse strain. At 26-38 weeks of age, female Fn14-knockout MRL-lpr/lpr mice had significantly lower levels of proteinuria compared with female wild-type MRL-lpr/lpr mice. Furthermore, Fn14-knockout mice had significantly improved renal histopathology accompanied by attenuated glomerular and tubulointerstitial inflammation. There was a significant reduction in glomerular Ig deposition in Fn14-knockout mice, despite no detectable differences in either serum levels of antibodies or splenic immune cell subsets. Notably, we found that the Fn14-knockout mice displayed substantial preservation of podocytes in glomeruli and that TWEAK signaling directly damaged barrier function and increased filtration through podocyte and glomerular endothelial cell monolayers. Our results show that deficiency of the Fn14 receptor significantly improves renal disease in a spontaneous lupus nephritis model through prevention of the direct injurious effects of TWEAK on the filtration barrier and/or modulation of cytokine production by resident kidney cells. Thus, blocking the TWEAK/Fn14 axis may be a novel therapeutic intervention in immune-mediated proliferative GN.


Asunto(s)
Factores de Crecimiento de Fibroblastos/deficiencia , Barrera de Filtración Glomerular/metabolismo , Nefritis Lúpica/etiología , Factores de Necrosis Tumoral/metabolismo , Animales , Citocina TWEAK , Femenino , Inmunoglobulina G/metabolismo , Ratones Noqueados , Proteinuria/metabolismo
2.
J Autoimmun ; 60: 40-50, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25911200

RESUMEN

Neuropsychiatric disease is one of the most common manifestations of human systemic lupus erythematosus, but the mechanisms remain poorly understood. In human brain microvascular endothelial cells in vitro, TNF-like weak inducer of apoptosis (TWEAK) decreases tight junction ZO-1 expression and increases the permeability of monolayer cell cultures. Furthermore, knockout (KO) of the TWEAK receptor, Fn14, in the MRL/lpr lupus mouse strain markedly attenuates neuropsychiatric disease, as demonstrated by significant reductions in depressive-like behavior and improved cognitive function. The purpose of the present study was to determine the mechanisms by which TWEAK signaling is instrumental in the pathogenesis of neuropsychiatric lupus (NPSLE). Evaluating brain sections of MRL/lpr Fn14WT and Fn14KO mice, we found that Fn14KO mice displayed significantly decreased cellular infiltrates in the choroid plexus. To evaluate the integrity of the blood brain barrier (BBB) in MRL/lpr mice, Western blot for fibronectin, qPCR for iNOS, and immunohistochemical staining for VCAM-1/ICAM-1 were performed. We found preserved BBB permeability in MRL/lpr Fn14KO mice, attributable to reduced brain expression of VCAM-1/ICAM-1 and iNOS. Additionally, administration of Fc-TWEAK intravenously directly increased the leakage of a tracer (dextran-FITC) into brain tissue. Furthermore, MRL/lpr Fn14KO mice displayed reduced antibody (IgG) and complement (C3, C6, and C4a) deposition in the brain. Finally, we found that MRL/lpr Fn14KO mice manifested reduced neuron degeneration and hippocampal gliosis. Our studies indicate that TWEAK/Fn14 interactions play an important role in the pathogenesis of NPSLE by increasing the accumulation of inflammatory cells in the choroid plexus, disrupting BBB integrity, and increasing neuronal damage, suggesting a novel target for therapy in this disease.


Asunto(s)
Apoptosis/genética , Barrera Hematoencefálica/fisiopatología , Neuronas/patología , Receptores del Factor de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Plexo Coroideo/fisiopatología , Cognición , Complemento C3/inmunología , Complemento C4a/inmunología , Complemento C6/inmunología , Citocina TWEAK , Depresión/genética , Modelos Animales de Enfermedad , Gliosis/genética , Inmunoglobulina G/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Permeabilidad , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor de TWEAK , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de la Zonula Occludens-1/biosíntesis
3.
Immunol Rev ; 244(1): 99-114, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22017434

RESUMEN

Our immune system performs the vital function of recognizing and eliminating invading pathogens and malignancies. There is an increasing appreciation that the immune system also actively mediates tissue responses under both physiological and pathological conditions, significantly impacting the inflammatory, fibrogenic, and regenerative components. Likewise, there is a growing understanding of how epithelial, endothelial, and other non-hematopoietic tissue cell types actively contribute to the interplay that shapes tissue responses. While much of the molecular basis underlying the immune regulation of tissue responses remains to be delineated, the tumor necrosis factor (TNF) superfamily ligand/receptor pair of TNF-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor-inducible molecule 14 (Fn14) has now emerged as a key piece of this puzzle. In this review, we first discuss how the usually 'dormant' TWEAK/Fn14 pathway becomes activated specifically in injury and disease contexts. We then summarize how TWEAK-mediated Fn14 signaling triggers a wide range of activities in tissue parenchymal and stromal cells as well as progenitor cells. Finally, we review recent experimental evidence that further supports the functional dichotomy of TWEAK/Fn14 activation in physiological versus pathological tissue responses and its potential therapeutic implications. Whereas transient TWEAK/Fn14 activation promotes productive tissue responses after injury, excessive or persistent TWEAK/Fn14 activation drives pathological tissue responses, leading to progressive damage and degeneration.


Asunto(s)
Inmunidad Innata , Inflamación/inmunología , Músculo Esquelético/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Células Madre/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Apoptosis/inmunología , Citocina TWEAK , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Expresión Génica/inmunología , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo
4.
J Exp Med ; 221(5)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38587494

RESUMEN

Jennifer S. Michaelson, Chief Scientific Officer at Cullinan Oncology, and Patrick A. Baeuerle, scientific advisor to Cullinan Oncology and honorary professor in immunology at Ludwig Maximilians University Munich, discuss the use of CD19-specific T cell-engaging antibody therapies (TCEs) as therapeutics for autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Humanos , Enfermedades Autoinmunes/terapia , Proteínas Adaptadoras Transductoras de Señales , Antígenos CD19 , Linfocitos T
5.
Cancer Immunol Res ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38842347

RESUMEN

Despite clinical evidence of antitumor activity, the development of cytokine therapies has been hampered by a narrow therapeutic window and limited response rates. Two cytokines of high interest for clinical development are interleukin 2 (IL-2) and interleukin 12 (IL-12), which potently synergize to promote the activation and proliferation of T cells and natural killer (NK) cells. However, the only approved human IL-2 therapy, Proleukin, is rarely used in the clinic due to systemic toxicities, and no IL-12 product has been approved to date due to severe dose-limiting toxicities. Here, we describe CLN-617, a first-in-class therapeutic for intratumoral (IT) injection that co-delivers IL-2 and IL-12 on a single molecule in a safe and effective manner. CLN-617 is a single-chain fusion protein comprised of IL-2, leukocyte-associated immunoglobulin-like receptor 2 (LAIR2), human serum albumin (HSA), and IL-12. LAIR2 and HSA function to retain CLN-617 in the treated tumor by binding collagen and increasing molecular weight, respectively. We found that IT administration of a murine surrogate of CLN-617, mCLN-617, eradicated established treated and untreated tumors in syngeneic models, significantly improved response to anti-PD1 checkpoint therapy, and generated a robust abscopal response dependent on cellular immunity and antigen cross-presentation. CLN-617 is being evaluated in a clinical trial in patients with advanced solid tumors (NCT06035744).

6.
J Autoimmun ; 43: 44-54, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23578591

RESUMEN

Given the early onset of neuropsychiatric disease and the potential response to immunosuppressive therapy, neuropsychiatric disease is considered a primary disease manifestation in systemic lupus erythematosus (SLE). However, the pathogenesis is not fully understood and optimal treatment has yet to be determined. TWEAK is a TNF family ligand that mediates pleotropic effects through its receptor Fn14, including the stimulation of inflammatory cytokine production by astrocytes, endothelial cells, and other non-hematopeotic cell types, and induction of neuronal death. Furthermore, TWEAK-inducible mediators are implicated in neuropsychiatric lupus. Thus, we hypothesized that the TWEAK/Fn14 pathway may be involved in the pathogenesis of neuropsychiatric SLE. We generated MRL-lpr/lpr (MRL/lpr) mice deficient for Fn14, the sole known signaling receptor for TWEAK. Neuropsychiatric disease was compared in age- and gender-matched MRL/lpr Fn14 wild type (WT) and knockout (KO) mice, using a comprehensive battery of neurobehavioral tests. We found that MRL/lpr Fn14WT mice displayed profound depression-like behavior as seen by increased immobility in a forced swim test and loss of preference for sweetened fluids, which were significantly ameliorated in Fn14KO mice. Similarly, MRL/lpr Fn14WT mice had impaired cognition, and this was significantly improved in Fn14KO mice. To determine the mechanism by which Fn14 deficiency ameliorates neuropsychiatric disease, we assessed the serum levels of autoantibodies and local expression of cytokines in the cortex and hippocampus of lupus mice. No significant differences were found in the serum levels of antibodies to nuclear antigens, or autoantibodies specifically associated with neuropsychiatric disease, between MRL/lpr Fn14WT and KO mice. However, MRL/lpr Fn14KO mice had significantly decreased brain expression of RANTES, C3, and other proinflammatory mediators. Furthermore, MRL/lpr Fn14KO mice displayed improved blood brain barrier integrity. In conclusion, several central manifestations of neuropsychiatric lupus, including depression-like behavior and altered cognition, are normalized in MRL/lpr mice lacking Fn14. Our results are the first to indicate a role for the TWEAK/Fn14 pathway in the pathogenesis of neuropsychiatric lupus, and suggest this ligand-receptor pair as a potential therapeutic target for a common and dangerous disease manifestation.


Asunto(s)
Vasculitis por Lupus del Sistema Nervioso Central/etiología , Vasculitis por Lupus del Sistema Nervioso Central/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Autoanticuerpos/sangre , Autoanticuerpos/líquido cefalorraquídeo , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Trastornos del Conocimiento/etiología , Citocina TWEAK , Depresión/etiología , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Vasculitis por Lupus del Sistema Nervioso Central/psicología , Ratones , Ratones Endogámicos MRL lpr , Ratones Noqueados , Neuroinmunomodulación , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal/inmunología , Receptor de TWEAK , Regulación hacia Arriba
7.
Cytokine ; 61(1): 210-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23107828

RESUMEN

The TWEAK receptor Fn14 (TNFRSF12), a member of the TNF Receptor superfamily, can mediate many processes, including apoptosis. Fn14 agonists have therefore been the subject of interest as potential cancer therapeutics. In cell culture experiments, interferon gamma (IFNγ) is typically required for induction of apoptotic activity by either TWEAK or Fn14 agonistic antibodies in most cell lines. We have investigated the mechanism of IFNγ signaling and the role of JAK-STAT signaling in TWEAK/Fn14-mediated tumor cell killing. We found that IFNγ-mediated enhancement of tumor cell killing is JAK-STAT dependent, as JAK inhibitors block IFNγ-dependent TWEAK induced apoptosis. Exposure of tumor cells to IFNγ results in an increase in Fn14 expression on the cell surface, which may be a mechanism by which IFNγ induces sensitivity to TWEAK. In a reciprocal fashion, we observed that IFNγ receptor levels increase in response to TWEAK treatment in WiDr cells. Significantly, we found that TWEAK alone can induce STAT1 phosphorylation in WiDr tumor cells. Moreover, TWEAK induction of tumor cell apoptosis in WiDr cells in the absence of IFNγ is mediated by the JAK-STAT pathway. Correspondingly, we show that treatment of tumor bearing mice with mBIIB036, an Fn14 agonistic antibody, results in STAT1 phosphorylation in the tumors. Notably, the level of STAT1 phosphorylation appears to correlate with the degree of tumor growth inhibition by BIIB036 in vivo. Additionally, in WiDr cells, TWEAK induces a soluble factor, which we have identified as IFNß, capable of independently inducing STAT1 phosphorylation when transferred to naïve cells. Finally, either IFNα or IFNß can partially substitute for IFNγ in sensitizing tumor cells to Fn14 agonists. In summary, we show that TWEAK/Fn14 can signal through the JAK-STAT pathway to induce IFNß, and that the ability of TWEAK to induce tumor cell apoptosis is mediated by JAK-STAT signaling. We also demonstrate that IFNγ enhancement of TWEAK/FN14-mediated tumor cell death is JAK-dependent and may occur by IFNγ-dependent upregulation of Fn14 on tumor cells. These findings may have implications for the appropriately targeted clinical development of Fn14 agonists as anti-cancer therapy.


Asunto(s)
Interferón gamma/metabolismo , Quinasas Janus/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Transcripción STAT1/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Interferón gamma/farmacología , Quinasas Janus/antagonistas & inhibidores , Ratones , Fosforilación , Unión Proteica , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Receptor de TWEAK
8.
Nat Cell Biol ; 8(8): 855-62, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16845383

RESUMEN

The tumour suppressor p53 induces apoptosis or cell-cycle arrest in response to genotoxic and other stresses. In unstressed cells, the anti-proliferative effects of p53 are restrained by mouse double minute 2 (Mdm2), a ubiquitin ligase (E3) that promotes p53 ubiquitination and degradation. Mdm2 also mediates its own degradation through auto-ubiquitination. It is unclear how the cis- and trans-E3 activities of Mdm2, which have opposing effects on cell fate, are differentially regulated. Here, we show that death domain-associated protein (Daxx) is required for Mdm2 stability. Downregulation of Daxx decreases Mdm2 levels, whereas overexpression of Daxx strongly stabilizes Mdm2. Daxx simultaneously binds to Mdm2 and the deubiquitinase Hausp, and it mediates the stabilizing effect of Hausp on Mdm2. In addition, Daxx enhances the intrinsic E3 activity of Mdm2 towards p53. On DNA damage, Daxx dissociates from Mdm2, which correlates with Mdm2 self-degradation. These findings reveal that Daxx modulates the function of Mdm2 at multiple levels and suggest that the disruption of the Mdm2-Daxx interaction may be important for p53 activation in response to DNA damage.


Asunto(s)
Proteínas Portadoras/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Animales , Western Blotting , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas Co-Represoras , Daño del ADN , Endopeptidasas/metabolismo , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HCT116 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Microscopía Fluorescente , Modelos Biológicos , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa , Peptidasa Específica de Ubiquitina 7
9.
J Immunother Cancer ; 11(8)2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37586770

RESUMEN

BACKGROUND: Despite significant progress in the development of T cell-engaging therapies for various B-cell malignancies, a high medical need remains for the refractory disease setting, often characterized by suboptimal target levels. METHODS: To address this issue, we have developed a 65-kDa multispecific antibody construct, CLN-978, with affinities tuned to optimize the killing of low-CD19 expressing tumor cells. CLN-978 bound to CD19 on B cells with picomolar affinity, and to CD3ε on T cells with nanomolar affinity. A serum albumin binding domain was incorporated to extend serum half-life. In this setting, we biophysically characterize and report the activities of CLN-978 in cell co-culture assays, multiple mouse models and non-human primates. RESULTS: Human T cells redirected by CLN-978 could eliminate target cells expressing less than 300 copies of CD19 on their surface. The half-life extension and high affinity for CD19 led to significant antitumor activity in murine lymphoma models at very low doses of CLN-978. In primates, we observed a long serum half-life, deep and sustained depletion of normal B cells, and remarkable tolerability, in particular, reduced cytokine release when CLN-978 was administered subcutaneously. CONCLUSIONS: CLN-978 warrants further exploration. An ongoing clinical phase 1 trial is investigating safety, pharmacokinetics, pharmacodynamics, and the initial therapeutic potential of subcutaneously administered CLN-978 in patients with non-Hodgkin's lymphoma.


Asunto(s)
Linfoma no Hodgkin , Neoplasias , Humanos , Animales , Ratones , Semivida , Proteínas Adaptadoras Transductoras de Señales , Anticuerpos , Antígenos CD19
10.
MAbs ; 15(1): 2208697, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37165468

RESUMEN

The field of immuno-oncology has revolutionized cancer patient care and improved survival and quality of life for patients. Much of the focus in the field has been on exploiting the power of the adaptive immune response through therapeutic targeting of T cells. While these approaches have markedly advanced the field, some challenges remain, and the clinical benefit of T cell therapies does not extend to all patients or tumor indications. Alternative strategies, such as engaging the innate immune system, have become an intense area of focus in the field. In particular, the engagement of natural killer (NK) cells as potent effectors of the innate immune response has emerged as a promising modality in immunotherapy. Here, we review therapeutic approaches for selective engagement of NK cells for cancer therapy, with a particular focus on targeting the key activating receptors NK Group 2D (NKG2D) and cluster of differentiation 16A (CD16A).


Asunto(s)
Subfamilia K de Receptores Similares a Lectina de Células NK , Neoplasias , Humanos , Calidad de Vida , Células Asesinas Naturales , Neoplasias/terapia , Inmunoterapia
11.
Clin Immunol ; 145(2): 108-21, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22982296

RESUMEN

Previously it was shown that the TNF superfamily member TWEAK (TNFSF12) acts through its receptor, Fn14, to promote proinflammatory responses in kidney cells, including the production of MCP-1, RANTES, IP-10 and KC. In addition, the TWEAK/Fn14 pathway promotes mesangial cell proliferation, vascular cell activation, and renal cell death. To study the relevance of the TWEAK/Fn14 pathway in the pathogenesis of antibody-induced nephritis using the mouse model of nephrotoxic serum nephritis (NTN), we induced NTN by passive transfer of rabbit anti-glomerular antibodies into Fn14 knockout (KO) and wild type (WT) mice. Severe proteinuria as well as renal histopathology were induced in WT but not in Fn14 KO mice. Similarly, a pharmacologic approach of anti-TWEAK mAb administration into WT mice in the NTN model significantly ameliorated proteinuria and improved kidney histology. Anti-TWEAK treatment did not affect the generation of mouse anti-rabbit antibodies; however, within the kidney there was a significant decrease in glomerular immunoglobulin deposition, as well as macrophage infiltrates and tubulointerstitial fibrosis. The mechanism of action is most likely due to reductions in downstream targets of TWEAK/Fn14 signaling, including reduced renal expression of MCP-1, VCAM-1, IP-10, RANTES as well as Fn14 itself, and other molecular pathways associated with fibrosis in anti-TWEAK treated mice. Thus, TWEAK/Fn14 interactions are instrumental in the pathogenesis of nephritis in the NTN model, apparently mediating a cascade of pathologic events locally in the kidney rather than by impacting the systemic immune response. Disrupting TWEAK/Fn14 interactions may be an innovative kidney-protective approach for the treatment of lupus nephritis and other antibody-induced renal diseases.


Asunto(s)
Anticuerpos/farmacología , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Riñón/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/inmunología , Inhibidores del Factor de Necrosis Tumoral , Animales , Anticuerpos/inmunología , Anticuerpos/uso terapéutico , Biomarcadores/metabolismo , Citocina TWEAK , Modelos Animales de Enfermedad , Fibrosis , Expresión Génica , Membrana Basal Glomerular/inmunología , Glomerulonefritis/terapia , Sueros Inmunes , Riñón/inmunología , Riñón/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Nefritis Lúpica/terapia , Células Mesangiales/efectos de los fármacos , Células Mesangiales/inmunología , Células Mesangiales/patología , Ratones , Ratones Noqueados , Conejos , Receptores del Factor de Necrosis Tumoral/deficiencia , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología
12.
J Autoimmun ; 39(3): 130-42, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22727560

RESUMEN

There is significant unmet need in the treatment of lupus nephritis (LN) patients. In this review, we highlight the role of the TWEAK/Fn14 pathway in mediating key pathologic processes underlying LN involving both glomerular and tubular injury, and thus the potential for renal protection via blockade of this pathway. The specific pathological mechanisms of TWEAK - namely promoting inflammation, renal cell proliferation and apoptosis, vascular activation and fibrosis - are described, with supporting data from animal models and in vitro systems. Furthermore, we detail the translational relevance of these mechanisms to clinical readouts in human LN. We present the opportunity for an anti-TWEAK therapeutic as a renal protective agent to improve efficacy relative to current standard of care treatments hopefully without increased safety risk, and highlight a phase II trial with BIIB023, an anti-TWEAK neutralizing antibody, designed to assess efficacy in LN patients. Taken together, targeting the TWEAK/Fn14 axis represents a potential new therapeutic paradigm for achieving renal protection in LN patients.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Glomérulos Renales/metabolismo , Túbulos Renales/metabolismo , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Inhibidores del Factor de Necrosis Tumoral , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Anticuerpos ampliamente neutralizantes , Proliferación Celular/efectos de los fármacos , Ensayos Clínicos Fase II como Asunto , Citocina TWEAK , Fibrosis , Expresión Génica/efectos de los fármacos , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Nefritis Lúpica/tratamiento farmacológico , Terapia Molecular Dirigida , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/metabolismo
13.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35288466

RESUMEN

BACKGROUND: In lymphoid malignancies, the introduction of chimeric antigen receptor T (CAR-T) cells and bispecific antibodies (bsAbs) has achieved remarkable clinical success. However, such immunotherapeutic strategies are not yet established for acute myeloid leukemia (AML), the most common form of acute leukemia in adults. Common targets in AML such as CD33, CD123, and CLEC12A are highly expressed on both AML blasts and on normal myeloid cells and hematopoietic stem cells (HSCs), thereby raising toxicity concerns. In B-cell acute lymphoblastic leukemia (B-ALL), bsAbs and CAR-T therapy targeting CD19 and CD22 have demonstrated clinical success, but resistance via antigen loss is common, motivating the development of agents focused on alternative targets. An attractive emerging target is FLT3, a proto-oncogene expressed in both AML and B-ALL, with low and limited expression on myeloid dendritic cells and HSCs. METHODS: We developed and characterized CLN-049, a T cell-activating bsAb targeting CD3 and FLT3, constructed as an IgG heavy chain/scFv fusion. CLN-049 binds the membrane proximal extracellular domain of the FLT3 protein tyrosine kinase, which facilitates the targeting of leukemic blasts regardless of FLT3 mutational status. CLN-049 was evaluated for preclinical safety and efficacy in vitro and in vivo. RESULTS: CLN-049 induced target-restricted activation of CD4+ and CD8+ T cells. AML cell lines expressing a broad range of surface levels of FLT3 were efficiently lysed on treatment with subnanomolar concentrations of CLN-049, whereas FLT3-expressing hematopoietic progenitor cells and dendritic cells were not sensitive to CLN-049 killing. Treatment with CLN-049 also induced lysis of AML and B-ALL patient blasts by autologous T cells at the low effector-to-target ratios typically observed in patients with overt disease. Lysis of leukemic cells was not affected by supraphysiological levels of soluble FLT3 or FLT3 ligand. In mouse xenograft models, CLN-049 was highly active against human leukemic cell lines and patient-derived AML and B-ALL blasts. CONCLUSIONS: CLN-049 has a favorable efficacy and safety profile in preclinical models, warranting evaluation of its antileukemic activity in the clinic.


Asunto(s)
Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Humanos , Inmunoglobulina G/uso terapéutico , Inmunoterapia Adoptiva , Subunidad alfa del Receptor de Interleucina-3 , Lectinas Tipo C , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Receptores Mitogénicos
14.
Circulation ; 119(15): 2058-68, 2009 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-19349318

RESUMEN

BACKGROUND: Tumor necrosis factor-like weak inducer of apoptosis (TWEAK), a member of the tumor necrosis factor superfamily, is a multifunctional cytokine known to regulate cellular functions in contexts of injury and disease through its receptor, fibroblast growth factor-inducible molecule 14 (Fn14). Although many of the processes and downstream signals regulated by the TWEAK/Fn14 pathway have been implicated in the development of cardiac dysfunction, the role of TWEAK in the cardiovascular system is completely unknown. METHODS AND RESULTS: Herein, we demonstrate that mouse and human cardiomyocytes express the TWEAK receptor Fn14. Furthermore, we determine that elevated circulating levels of TWEAK, induced via transgenic or adenoviral-mediated gene expression in mice, result in dilated cardiomyopathy with subsequent severe cardiac dysfunction. This phenotype was mediated exclusively by the Fn14 receptor, independent of tumor necrosis factor-alpha, and was associated with cardiomyocyte elongation and cardiac fibrosis but not cardiomyocyte apoptosis. Moreover, we find that circulating TWEAK levels were differentially upregulated in patients with idiopathic dilated cardiomyopathy compared with other forms of heart disease and normal control subjects. CONCLUSIONS: Our data suggest that TWEAK/Fn14 may be important in regulating myocardial structural remodeling and function and may play a role in the pathogenesis of dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Factores de Necrosis Tumoral/fisiología , Animales , Apoptosis , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Tamaño de la Célula , Enfermedad Coronaria/metabolismo , Enfermedad Coronaria/patología , Citocina TWEAK , Femenino , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Humanos , Hipertensión/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Persona de Mediana Edad , Fenotipo , Proteínas Recombinantes de Fusión/fisiología , Receptor de TWEAK , Transducción Genética , Factores de Necrosis Tumoral/sangre , Factores de Necrosis Tumoral/genética
15.
Gastroenterology ; 136(3): 912-23, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19109961

RESUMEN

BACKGROUND AND AIMS: Tumor necrosis factor (TNF) superfamily members have attracted attention as new therapeutic targets for treating inflammatory disease. TNF-like weak inducer of apoptosis (TWEAK) is a unique, multifunctional TNF family cytokine that signals through its receptor, fibroblast growth factor-inducible molecule 14 (Fn14). The role of this pathway in the intestine has not been previously reported. METHODS: The 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis model was conducted in TWEAK- or Fn14-deficient mice or in normal mice treated with a TWEAK-blocking monoclonal antibody, and clinical severity, histopathology, immunohistochemistry for cell infiltrates, TWEAK and Fn14, gene expression profiling in the colon, and systemic adaptive immunity were assessed. The effect of TWEAK on colon epithelial cell production of inflammatory mediators was analyzed in vitro. The gamma-irradiation injury model was conducted in TWEAK- or Fn14-deficient mice, and crypt epithelial death was assessed. RESULTS: Colitis severity and histologic scores were significantly reduced by TWEAK pathway deficiency or TWEAK-blocking monoclonal antibody. Neutrophil and macrophage infiltrates, chemokines, cytokines, and matrix metalloproteinase expression were reduced in the TWEAK-deficient colon after TNBS administration; however, systemic adaptive immune responses to trinitrophenyl were not altered. Fn14 is expressed on colon epithelial cells in TNBS colitis, and TWEAK induces epithelial production of pathogenic mediators. TWEAK also regulates intestinal epithelial turnover, as evidenced by reduced epithelial cell death after gamma-irradiation injury in TWEAK and Fn14 knockout mice. CONCLUSIONS: Our studies elucidate a nonredundant TWEAK-intestinal epithelial cell axis and suggest that blocking TWEAK may dampen chronic intestinal inflammation and allow normal epithelial repair.


Asunto(s)
Colitis/metabolismo , Colitis/patología , Mucosa Intestinal/patología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Colitis/inmunología , Colon/inmunología , Colon/metabolismo , Colon/patología , Citocina TWEAK , Modelos Animales de Enfermedad , Rayos gamma , Sistema Inmunológico/fisiología , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Neutrófilos/patología , ARN Mensajero/metabolismo , Receptor de TWEAK , Factores de Necrosis Tumoral/genética , Úlcera/inmunología , Úlcera/metabolismo , Úlcera/patología
16.
Cytokine ; 46(1): 24-35, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19233685

RESUMEN

Members of the TNF-ligand and receptor superfamilies are important in the pathogenesis of lupus nephritis, a major cause of mortality and morbidity in SLE. TWEAK, a member of the TNF-ligand superfamily, is markedly increased in urine from patients with active lupus nephritis, and urinary TWEAK levels significantly correlate with renal disease activity. To support a possible role of TWEAK in the pathogenesis of lupus nephritis and other inflammatory nephritides, we examined the effects of TWEAK in human kidney mesangial cells, podocytes and tubular cells, following our demonstration of the presence of the TWEAK receptor Fn14 on these cells. We found that TWEAK induces human kidney cells to express multiple inflammatory mediators, including RANTES, MCP-1, IP-10, MIP-1alpha, ICAM-1, and VCAM-1. Cytokine production is mediated through NF-kappaB activation, and is inhibited by anti-TWEAK monoclonal antibodies. TWEAK stimulated chemokines induced migration of human PBMC, particularly monocytes/macrophages. Furthermore, we found that TWEAK promotes kidney infiltration of inflammatory cells, and stimulates proliferation of kidney cells in vitro and in vivo. Thus, TWEAK may play an important pathogenic role in the development of glomerulonephritis by promoting a local inflammatory environment and inducing kidney cell proliferation. Blocking TWEAK/Fn14 interactions may be a promising therapeutic target in immune-mediated renal diseases.


Asunto(s)
Riñón/citología , Riñón/metabolismo , Receptores del Factor de Necrosis Tumoral/fisiología , Animales , Proliferación Celular , Citocinas/metabolismo , Glomerulonefritis/metabolismo , Humanos , Inflamación , Leucocitos Mononucleares/citología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Receptor de TWEAK
17.
J Clin Invest ; 115(9): 2330-40, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16110324

RESUMEN

Progenitor ("oval") cell expansion accompanies many forms of liver injury, including alcohol toxicity and submassive parenchymal necrosis as well as experimental injury models featuring blocked hepatocyte replication. Oval cells can potentially become either hepatocytes or biliary epithelial cells and may be critical to liver regeneration, particularly when hepatocyte replication is impaired. The regulation of oval cell proliferation is incompletely understood. Herein we present evidence that a TNF family member called TWEAK (TNF-like weak inducer of apoptosis) stimulates oval cell proliferation in mouse liver through its receptor Fn14. TWEAK has no effect on mature hepatocytes and thus appears to be selective for oval cells. Transgenic mice overexpressing TWEAK in hepatocytes exhibit periportal oval cell hyperplasia. A similar phenotype was obtained in adult wild-type mice, but not Fn14-null mice, by administering TWEAK-expressing adenovirus. Oval cell expansion induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) was significantly reduced in Fn14-null mice as well as in adult wild-type mice with a blocking anti-TWEAK mAb. Importantly, TWEAK stimulated the proliferation of an oval cell culture model. Finally, we show increased Fn14 expression in chronic hepatitis C and other human liver diseases relative to its expression in normal liver, which suggests a role for the TWEAK/Fn14 pathway in human liver injury. We conclude that TWEAK has a selective mitogenic effect for liver oval cells that distinguishes it from other previously described growth factors.


Asunto(s)
Proliferación Celular , Hígado/citología , Hígado/metabolismo , Células Madre/fisiología , Factores de Necrosis Tumoral/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Citocina TWEAK , Femenino , Humanos , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/citología , Receptor de TWEAK , Distribución Tisular , Transgenes , Factores de Necrosis Tumoral/genética
18.
J Cereb Blood Flow Metab ; 27(3): 534-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16835630

RESUMEN

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor superfamily. TWEAK acts via binding to a cell surface receptor named Fn14. To study the role of this cytokine in the regulation of the permeability of the neurovascular unit (NVU) during cerebral ischemia, TWEAK activity was inhibited in wild-type mice with a soluble Fn14-Fc decoy receptor administered either immediately or 1 h after middle cerebral artery occlusion (MCAO). Administration of Fn14-Fc decoy resulted in faster recovery of motor function and a 66.4%+/-10% decrease in Evans blue dye extravasation when treatment was administered immediately after MCAO and a 46.1%+/-13.1% decrease when animals were treated 1 h later (n=4, P<0.05). Genetic deficiency of Fn14 resulted in a 60%+/-12.8% decrease in the volume of the ischemic lesion (n=6, P<0.05), and a 87%+/-22% inhibition in Evans blue dye extravasation 48 h after the onset of the ischemic insult (n=6, P<0.005). Compared with control animals, treatment with Fn14-Fc decoy or genetic deficiency of Fn14 also resulted in a significant inhibition of nuclear factor-kappaB pathway activation, matrix metalloproteinase-9 activation and basement membrane laminin degradation after MCAO. These findings show that the cytokine TWEAK plays a role in the disruption of the structure of the NVU during cerebral ischemia and that TWEAK antagonism is a potential therapeutic strategy for acute cerebral ischemia.


Asunto(s)
Barrera Hematoencefálica/ultraestructura , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Permeabilidad Capilar/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/ultraestructura , Barrera Hematoencefálica/metabolismo , Western Blotting , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Edema Encefálico , Isquemia Encefálica/etiología , Citocina TWEAK , Ensayo de Cambio de Movilidad Electroforética , Células Endoteliales/ultraestructura , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz , Ratones , Microscopía Electrónica de Transmisión , FN-kappa B/metabolismo , Recuperación de la Función , Receptor de TWEAK
19.
Cytokine ; 40(1): 1-16, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17981048

RESUMEN

First described as a weak apoptosis inducer, the TNF superfamily ligand TWEAK has since emerged as a cytokine that regulates multiple cellular responses, including proinflammatory activity, angiogenesis and cell proliferation, suggesting roles in inflammation and cancer. More recently TWEAK's ability to regulate progenitor cell fate was elucidated. Experiments using genetic overexpression and pathway inhibition or deficiency in mice indicate that TWEAK coordinates inflammatory and progenitor cell responses in settings of acute injury through its highly inducible receptor, FGF-inducible molecule 14 (Fn14), establishing the pathway's physiological role in facilitating acute tissue repair. In contrast, in chronic inflammatory disease models characterized by persistent TWEAK/Fn14 activation, TWEAK functions as a novel pathogenic mediator by amplifying inflammation, promoting tissue damage and potentially impeding endogenous repair mechanisms. Herein we aim not only to review the multifaceted functions of this emerging pathway, but also propose a conceptual framework for TWEAK/Fn14 pathway function in health and disease, supported by studies employing TWEAK and Fn14 deficient mice and anti-TWEAK blocking mAbs in acute injury and inflammatory disease settings. In addition to a perspective of the biology, we discuss potential therapeutic strategies targeting this pathway for the treatment of tissue injury, chronic inflammatory diseases and cancer.


Asunto(s)
Citocinas/fisiología , Mediadores de Inflamación/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Transducción de Señal/fisiología , Factores de Necrosis Tumoral/fisiología , Cicatrización de Heridas/fisiología , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Citocina TWEAK , Humanos , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/genética , Receptor de TWEAK , Factores de Necrosis Tumoral/biosíntesis , Cicatrización de Heridas/genética
20.
Ann N Y Acad Sci ; 1109: 265-74, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17785315

RESUMEN

Lupus nephritis (LN) is a major cause of morbidity and mortality in systemic lupus erythematosus (SLE). As the course of LN is often unpredictable, it is important to identify reliable, noninvasive methods to repeatedly assess the condition of the kidneys in these patients. Urinary biomarkers are easily obtained and probably are best at reflecting the current renal status, as they specifically represent local inflammatory activity. In this article, we review a number of potential urinary biomarkers, including Lipocalin-2, MCP-1, IP-10, IL-6, and IL-8. Additionally, we provide evidence for the possible utility of the novel cytokine TWEAK as a urinary biomarker for LN.


Asunto(s)
Proteínas de Fase Aguda/orina , Citocinas/orina , Nefritis Lúpica/orina , Proteínas Proto-Oncogénicas/orina , Animales , Biomarcadores/orina , Estudios de Seguimiento , Humanos , Lipocalina 2 , Lipocalinas , Nefritis Lúpica/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA