Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
bioRxiv ; 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38464306

RESUMEN

Sterile tissue injury, such as by acute kidney injury, is common in the clinic and frequently associated with respiratory compromise and hypoxemia. We previously described signaling components released by the injured kidney that drive a remote inflammatory response in the lung. How this caused the resultant hypoxemia remained unclear. Here, we report that sterile kidney tissue injury induces rapid intravascular "neutrophil train" formation in lung capillaries, a novel form of neutrophil swarming. Rapid swarming is enhanced by decreased deformability of circulating neutrophils that impedes their lung capillary passage. Classical lung monocytes are required for neutrophil train formation and release CXCL2 to attract and retain stiffened neutrophils in lung capillaries which reduces capillary perfusion. We thus discovered a novel feature of kidney-lung crosstalk after sterile kidney tissue injury, capillary perfusion deficits that lead to reduced oxygenation despite proper alveolar function and ventilation, unlike in infectious inflammatory lung processes, such as bacterial pneumonia.

2.
Nat Rev Nephrol ; 20(2): 120-136, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37667081

RESUMEN

The homeostasis and health of an organism depend on the coordinated interaction of specialized organs, which is regulated by interorgan communication networks of circulating soluble molecules and neuronal connections. Many diseases that seemingly affect one primary organ are really multiorgan diseases, with substantial secondary remote organ complications that underlie a large part of their morbidity and mortality. Acute kidney injury (AKI) frequently occurs in critically ill patients with multiorgan failure and is associated with high mortality, particularly when it occurs together with respiratory failure. Inflammatory lung lesions in patients with kidney failure that could be distinguished from pulmonary oedema due to volume overload were first reported in the 1930s, but have been largely overlooked in clinical settings. A series of studies over the past two decades have elucidated acute and chronic kidney-lung and lung-kidney interorgan communication networks involving various circulating inflammatory cytokines and chemokines, metabolites, uraemic toxins, immune cells and neuro-immune pathways. Further investigations are warranted to understand these clinical entities of high morbidity and mortality, and to develop effective treatments.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Respiratoria , Humanos , Riñón , Pulmón , Lesión Renal Aguda/complicaciones , Citocinas , Enfermedad Crítica
3.
Cells ; 12(18)2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37759437

RESUMEN

Elevated levels of circulating tumor necrosis factor receptors 1 and 2 (cTNFR1/2) predict chronic kidney disease (CKD) progression; however, the mechanisms of their release remain unknown. Whether acute kidney injury (AKI) drives cTNFR1/2 elevations and whether they predict disease outcomes after AKI remain unknown. In this study, we used AKI patient serum and urine samples, mouse models of kidney injury (ischemic, obstructive, and toxic), and progression to fibrosis, nephrectomy, and related single-cell RNA-sequencing datasets to experimentally test the role of kidney injury on cTNFR1/2 levels. We show that TNFR1/2 serum and urine levels are highly elevated in all of the mouse models of kidney injury tested, beginning within one hour post injury, and correlate with its severity. Consistent with this, serum and urine TNFR1/2 levels are increased in AKI patients and correlate with the severity of kidney failure. Kidney tissue expression of TNFR1/2 after AKI is only slightly increased and bilateral nephrectomies lead to strong cTNFR1/2 elevations, suggesting the release of these receptors by extrarenal sources. The injection of the uremic toxin indoxyl sulfate in healthy mice induces moderate cTNFR1/2 elevations. Moreover, TNF neutralization does not affect early cTNFR1/2 elevations after AKI. These data suggest that cTNFR1/2 levels in AKI do not reflect injury-induced TNF activity, but rather a rapid response to loss of kidney function and uremia. In contrast to traditional disease biomarkers, such as serum creatinine or BUN, cTNFR1/2 levels remain elevated for weeks after severe kidney injury. At these later timepoints, cTNFR1/2 levels positively correlate with remaining kidney injury. During the AKI-to-CKD transition, elevations of TNFR1/2 kidney expression and of cTNFR2 levels correlate with kidney fibrosis levels. In conclusion, our data demonstrate that kidney injury drives acute increases in cTNFR1/2 serum levels, which negatively correlate with kidney function. Sustained TNFR1/2 elevations after kidney injury during AKI-to-CKD transition reflect persistent tissue injury and progression to kidney fibrosis.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Humanos , Animales , Ratones , Receptores Tipo I de Factores de Necrosis Tumoral , Riñón , Modelos Animales de Enfermedad , Fibrosis
4.
Nephrol Dial Transplant ; 38(5): 1139-1150, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36269313

RESUMEN

BACKGROUND: Inflammation is a key driver of the transition of acute kidney injury to progressive fibrosis and chronic kidney disease (AKI-to-CKD transition). Blocking a-disintegrin-and-metalloprotease-17 (ADAM17)-dependent ectodomain shedding, in particular of epidermal growth factor receptor (EGFR) ligands and of the type 1 inflammatory cytokine tumor necrosis factor (TNF), reduces pro-inflammatory and pro-fibrotic responses after ischemic AKI or unilateral ureteral obstruction (UUO), a classical fibrosis model. Metalloprotease or EGFR inhibition show significant undesirable side effects in humans. In retrospective studies anti-TNF biologics reduce the incidence and progression of CKD in humans. Whether TNF has a role in AKI-to-CKD transition and how TNF inhibition compares to EGFR inhibition is largely unknown. METHODS: Mice were subjected to bilateral renal ischemia-reperfusion injury or unilateral ureteral obstruction. Kidneys were analyzed by histology, immunohistochemistry, qPCR, western blot, mass cytometry, scRNA sequencing, and cytokine profiling. RESULTS: Here we show that TNF or EGFR inhibition reduce AKI-to-CKD transition and fibrosis equally by about 25%, while combination has no additional effect. EGFR inhibition reduced kidney TNF expression by about 50% largely by reducing accumulation of TNF expressing immune cells in the kidney early after AKI, while TNF inhibition did not affect EGFR activation or immune cell accumulation. Using scRNAseq data we show that TNF is predominantly expressed by immune cells in AKI but not in proximal tubule cells (PTC), and PTC-TNF knockout did not affect AKI-to-CKD transition in UUO. Thus, the anti-inflammatory and anti-fibrotic effects of the anti-TNF biologic etanercept in AKI-to-CKD transition rely on blocking TNF that is released from immune cells recruited or accumulating in response to PTC-EGFR signals. CONCLUSION: Short-term anti-TNF biologics during or after AKI could be helpful in the prevention of AKI-to-CKD transition.


Asunto(s)
Lesión Renal Aguda , Productos Biológicos , Insuficiencia Renal Crónica , Obstrucción Ureteral , Humanos , Ratones , Animales , Etanercept/farmacología , Etanercept/uso terapéutico , Etanercept/metabolismo , Obstrucción Ureteral/metabolismo , Estudios Retrospectivos , Inhibidores del Factor de Necrosis Tumoral/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacología , Insuficiencia Renal Crónica/patología , Riñón/patología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Receptores ErbB , Factor de Necrosis Tumoral alfa/metabolismo , Fibrosis , Productos Biológicos/metabolismo , Productos Biológicos/farmacología
6.
Sci Adv ; 8(8): eabm5900, 2022 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-35213222

RESUMEN

Tissue injury can drive secondary organ injury; however, mechanisms and mediators are not well understood. To identify interorgan cross-talk mediators, we used acute kidney injury (AKI)-induced acute lung injury (ALI) as a clinically important example. Using kidney and lung single-cell RNA sequencing after AKI in mice followed by ligand-receptor pairing analysis across organs, kidney ligands to lung receptors, we identify kidney-released circulating osteopontin (OPN) as a novel AKI-ALI mediator. OPN release from kidney tubule cells triggered lung endothelial leakage, inflammation, and respiratory failure. Pharmacological or genetic OPN inhibition prevented AKI-ALI. Transplantation of ischemic wt kidneys caused AKI-ALI, but not of ischemic OPN-global knockout kidneys, identifying kidney-released OPN as necessary interorgan signal to cause AKI-ALI. We show that OPN serum levels are elevated in patients with AKI and correlate with kidney injury. Our results demonstrate feasibility of using ligand-receptor analysis across organs to identify interorgan cross-talk mediators and may have important therapeutic implications in human AKI-ALI and multiorgan failure.


Asunto(s)
Lesión Renal Aguda , Lesión Pulmonar Aguda , Insuficiencia Respiratoria , Lesión Renal Aguda/etiología , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/prevención & control , Animales , Femenino , Humanos , Riñón , Ligandos , Masculino , Ratones , Osteopontina
7.
FEBS Lett ; 596(5): 620-637, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34932216

RESUMEN

Homoeostasis and health of multicellular organisms with multiple organs depends on interorgan communication. Tissue injury in one organ disturbs this homoeostasis and can lead to disease in multiple organs, or multiorgan failure. Many routes of interorgan crosstalk during homoeostasis are relatively well known, but interorgan crosstalk in disease still lacks understanding. In particular, how tissue injury in one organ can drive injury at remote sites and trigger multiorgan failure with high mortality is poorly understood. As examples, acute kidney injury can trigger acute lung injury and cardiovascular dysfunction; pneumonia, sepsis or liver failure conversely can cause kidney failure; lung transplantation very frequently triggers acute kidney injury. Mechanistically, interorgan crosstalk after tissue injury could involve soluble mediators and their target receptors, cellular mediators, in particular immune cells, as well as newly identified neuro-immune connections. In this review, I will focus the discussion of deleterious interorgan crosstalk and its mechanistic concepts on one example, acute kidney injury-induced remote lung injury.


Asunto(s)
Lesión Renal Aguda , Lesión Pulmonar Aguda , Sepsis , Lesión Renal Aguda/etiología , Lesión Pulmonar Aguda/etiología , Femenino , Humanos , Riñón , Pulmón , Masculino , Insuficiencia Multiorgánica/etiología , Sepsis/complicaciones
8.
J Clin Invest ; 129(12): 5501-5517, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31710314

RESUMEN

The proximal tubule has a remarkable capacity for repair after acute injury, but the cellular lineage and molecular mechanisms underlying this repair response are incompletely understood. Here, we developed a Kim1-GFPCreERt2 knockin mouse line (Kim1-GCE) in order to perform genetic lineage tracing of dedifferentiated cells while measuring the cellular transcriptome of proximal tubule during repair. Acutely injured genetically labeled clones coexpressed KIM1, VIMENTIN, SOX9, and KI67, indicating a dedifferentiated and proliferative state. Clonal analysis revealed clonal expansion of Kim1+ cells, indicating that acutely injured, dedifferentiated proximal tubule cells, rather than fixed tubular progenitor cells, account for repair. Translational profiling during injury and repair revealed signatures of both successful and unsuccessful maladaptive repair. The transcription factor Foxm1 was induced early in injury, was required for epithelial proliferation in vitro, and was dependent on epidermal growth factor receptor (EGFR) stimulation. In conclusion, dedifferentiated proximal tubule cells effect proximal tubule repair, and we reveal an EGFR/FOXM1-dependent signaling pathway that drives proliferative repair after injury.


Asunto(s)
Lesión Renal Aguda/patología , Proteína Forkhead Box M1/fisiología , Túbulos Renales Proximales/patología , Daño por Reperfusión/patología , Adulto , Animales , Desdiferenciación Celular , Linaje de la Célula , Proliferación Celular , Modelos Animales de Enfermedad , Receptores ErbB/fisiología , Femenino , Humanos , Riñón/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad
9.
J Am Soc Nephrol ; 30(12): 2370-2383, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31676723

RESUMEN

BACKGROUND: Sustained activation of EGF receptor (EGFR) in proximal tubule cells is a hallmark of progressive kidney fibrosis after AKI and in CKD. However, the molecular mechanisms and particular EGFR ligands involved are unknown. METHODS: We studied EGFR activation in proximal tubule cells and primary tubular cells isolated from injured kidneys in vitro. To determine in vivo the role of amphiregulin, a low-affinity EGFR ligand that is highly upregulated with injury, we used ischemia-reperfusion injury or unilateral ureteral obstruction in mice with proximal tubule cell-specific knockout of amphiregulin. We also injected soluble amphiregulin into knockout mice with proximal tubule cell-specific deletion of amphiregulin's releasing enzyme, the transmembrane cell-surface metalloprotease, a disintegrin and metalloprotease-17 (ADAM17), and into ADAM17 hypomorphic mice. RESULTS: Yes-associated protein 1 (YAP1)-dependent upregulation of amphiregulin transcript and protein amplifies amphiregulin signaling in a positive feedback loop. YAP1 also integrates signals of other moderately injury-upregulated, low-affinity EGFR ligands (epiregulin, epigen, TGFα), which also require soluble amphiregulin and YAP1 to induce sustained EGFR activation in proximal tubule cells in vitro. In vivo, soluble amphiregulin injection sufficed to reverse protection from fibrosis after ischemia-reperfusion injury in ADAM17 hypomorphic mice; injected soluble amphiregulin also reversed the corresponding protective proximal tubule cell phenotype in injured proximal tubule cell-specific ADAM17 knockout mice. Moreover, the finding that proximal tubule cell-specific amphiregulin knockout mice were protected from fibrosis after ischemia-reperfusion injury or unilateral ureteral obstruction demonstrates that amphiregulin was necessary for the development of fibrosis. CONCLUSIONS: Our results identify amphiregulin as a key player in injury-induced kidney fibrosis and suggest therapeutic or diagnostic applications of soluble amphiregulin in kidney disease.


Asunto(s)
Lesión Renal Aguda/metabolismo , Anfirregulina/fisiología , Receptores ErbB/agonistas , Túbulos Renales Proximales/metabolismo , Insuficiencia Renal Crónica/patología , Proteína ADAM17/deficiencia , Proteína ADAM17/genética , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/patología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Anfirregulina/deficiencia , Animales , Proteínas de Ciclo Celular/fisiología , Células Cultivadas , Familia de Proteínas EGF/metabolismo , Células Epiteliales/metabolismo , Fibrosis , Riñón/irrigación sanguínea , Masculino , Ratones , Ratones Noqueados , Procesamiento Proteico-Postraduccional , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Regulación hacia Arriba , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología , Proteínas Señalizadoras YAP
11.
Mol Cancer Res ; 16(1): 147-161, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29018056

RESUMEN

Ectodomain shedding of cell-surface precursor proteins by metalloproteases generates important cellular signaling molecules. Of importance for disease is the release of ligands that activate the EGFR, such as TGFα, which is mostly carried out by ADAM17 [a member of the A-disintegrin and metalloprotease (ADAM) domain family]. EGFR ligand shedding has been linked to many diseases, in particular cancer development, growth and metastasis, as well as resistance to cancer therapeutics. Excessive EGFR ligand release can outcompete therapeutic EGFR inhibition or the inhibition of other growth factor pathways by providing bypass signaling via EGFR activation. Drugging metalloproteases directly have failed clinically because it indiscriminately affected shedding of numerous substrates. It is therefore essential to identify regulators for EGFR ligand cleavage. Here, integration of a functional shRNA genomic screen, computational network analysis, and dedicated validation tests succeeded in identifying several key signaling pathways as novel regulators of TGFα shedding in cancer cells. Most notably, a cluster of genes with NFκB pathway regulatory functions was found to strongly influence TGFα release, albeit independent of their NFκB regulatory functions. Inflammatory regulators thus also govern cancer cell growth-promoting ectodomain cleavage, lending mechanistic understanding to the well-known connection between inflammation and cancer.Implications: Using genomic screens and network analysis, this study defines targets that regulate ectodomain shedding and suggests new treatment opportunities for EGFR-driven cancers. Mol Cancer Res; 16(1); 147-61. ©2017 AACR.


Asunto(s)
Neoplasias/genética , Neoplasias/metabolismo , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo , Línea Celular Tumoral , Receptores ErbB/metabolismo , Redes Reguladoras de Genes , Genómica/métodos , Humanos , Células Jurkat , Ligandos , Modelos Genéticos , ARN Interferente Pequeño/genética , Transducción de Señal
12.
Trends Cancer ; 3(7): 482-490, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28718403

RESUMEN

Identification of early cancer, detection of progression, and monitoring of therapeutic success remain urgent issues in clinical medicine, particularly given the increasing cancer incidence in our aging populations. New methodologies have provided enormous progress over the past decades by defining the genetic and proteomic composition of cancers, yielding putative cancer biomarkers detectable in blood or other body fluids less invasively and more cheaply than using currently available screening techniques that often involve biopsies or surgery. However, the clinical use of these new methodologies is still far off. In this review, we focus on putative soluble cancer biomarkers shed from the cell surface by metalloproteases overexpressed in numerous cancers. Although useful candidates have been identified, their validation and adoption into clinical use remain challenging.


Asunto(s)
Proteínas ADAM/metabolismo , Biomarcadores de Tumor/análisis , Detección Precoz del Cáncer/métodos , Proteínas de Neoplasias/análisis , Neoplasias/diagnóstico , Biomarcadores de Tumor/metabolismo , Líquidos Corporales/química , Membrana Celular/metabolismo , Monitoreo de Drogas/métodos , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/patología , Péptidos/análisis , Péptidos/metabolismo , Proteolisis , Proteómica/métodos
13.
Sci Rep ; 6: 37464, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27876763

RESUMEN

Release of cytokines, growth factors and other life-essential molecules from precursors by a-disintegrin-and-metalloproteases (ADAMs) is regulated with high substrate-specificity. We hypothesized that this is achieved by cleavage-regulatory intracellular-domain (ICD)-modifications of the precursors. We show here that cleavage-stimuli-induced specific ICD-modifications cause structural substrate changes that enhance ectodomain sensitivity of neuregulin-1 (NRG1; epidermal-growth-factor) or CD44 (receptor-tyrosine-kinase (RTK) co-receptor) to chymotrypsin/trypsin or soluble ADAM. This inside-out signal transfer required substrate homodimerization and was prevented by cleavage-inhibitory ICD-mutations. In chimeras, regulation could be conferred to a foreign ectodomain, suggesting a common higher-order structure. We predict that substrate-specific protease-accessibility-regulation controls release of numerous ADAM substrates.


Asunto(s)
Proteínas ADAM/genética , Receptores de Hialuranos/genética , Neurregulina-1/genética , Proteínas ADAM/química , Animales , Quimera/genética , Receptores de Hialuranos/química , Ratones , Mutación , Células 3T3 NIH , Neurregulina-1/química , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Dominios Proteicos/genética , Especificidad por Sustrato/genética
14.
JCI Insight ; 1(13)2016 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-27642633

RESUMEN

Kidney fibrosis following kidney injury is an unresolved health problem and causes significant morbidity and mortality worldwide. In a study into its molecular mechanism, we identified essential causative features. Acute or chronic kidney injury causes sustained elevation of a disintegrin and metalloprotease 17 (ADAM17); of its cleavage-activated proligand substrates, in particular of pro-TNFα and the EGFR ligand amphiregulin (pro-AREG); and of the substrates' receptors. As a consequence, EGFR is persistently activated and triggers the synthesis and release of proinflammatory and profibrotic factors, resulting in macrophage/neutrophil ingress and fibrosis. ADAM17 hypomorphic mice, specific ADAM17 inhibitor-treated WT mice, or mice with inducible KO of ADAM17 in proximal tubule (Slc34a1-Cre) were significantly protected against these effects. In vitro, in proximal tubule cells, we show that AREG has unique profibrotic actions that are potentiated by TNFα-induced AREG cleavage. In vivo, in acute kidney injury (AKI) and chronic kidney disease (CKD, fibrosis) patients, soluble AREG is indeed highly upregulated in human urine, and both ADAM17 and AREG expression show strong positive correlation with fibrosis markers in related kidney biopsies. Our results indicate that targeting of the ADAM17 pathway represents a therapeutic target for human kidney fibrosis.


Asunto(s)
Proteína ADAM17/metabolismo , Lesión Renal Aguda/patología , Anfirregulina/farmacología , Túbulos Renales Proximales/metabolismo , Riñón/patología , Lesión Renal Aguda/metabolismo , Animales , Células Cultivadas , Receptores ErbB/metabolismo , Fibrosis , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb
15.
Mol Cell Biol ; 35(19): 3381-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26217011

RESUMEN

Ectodomain cleavage by A-disintegrin and -metalloproteases (ADAMs) releases many important biologically active substrates and is therefore tightly controlled. Part of the regulation occurs on the level of the enzymes and affects their cell surface abundance and catalytic activity. ADAM-dependent proteolysis occurs outside the plasma membrane but is mostly controlled by intracellular signals. However, the intracellular domains (ICDs) of ADAM10 and -17 can be removed without consequences for induced cleavage, and so far it is unclear how intracellular signals address cleavage. We therefore explored whether substrates themselves could be chosen for proteolysis via ICD modification. We report here that CD44 (ADAM10 substrate), a receptor tyrosine kinase (RTK) coreceptor required for cellular migration, and pro-NRG1 (ADAM17 substrate), which releases the epidermal growth factor (EGF) ligand neuregulin required for axonal outgrowth and myelination, are indeed posttranslationally modified at their ICDs. Tetradecanoyl phorbol acetate (TPA)-induced CD44 cleavage requires dephosphorylation of ICD serine 291, while induced neuregulin release depends on the phosphorylation of several NRG1-ICD serines, in part mediated by protein kinase Cδ (PKCδ). Downregulation of PKCδ inhibits neuregulin release and reduces ex vivo neurite outgrowth and myelination of trigeminal ganglion explants. Our results suggest that specific selection among numerous substrates of a given ADAM is determined by ICD modification of the substrate.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Técnicas de Cocultivo , Células HEK293 , Humanos , Receptores de Hialuranos/química , Datos de Secuencia Molecular , Neurregulina-1/química , Neuritas/fisiología , Fosforilación , Proteína Quinasa C-delta/metabolismo , Proteolisis , Células de Schwann/metabolismo , Transducción de Señal , Ganglio del Trigémino/citología
16.
J Biol Chem ; 290(28): 17041-54, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25925953

RESUMEN

Ectodomain shedding of transmembrane precursor proteins generates numerous life-essential molecules, such as epidermal growth factor receptor ligands. This cleavage not only releases the regulatory growth factor, but it is also the required first step for the subsequent processing by γ-secretase and the release of gene regulatory intracellular fragments. Signaling within the cell modifies the cytoplasmic tails of substrates, a step important in starting the specific and regulated cleavage of a large number of studied substrates. Ectodomain cleavage occurs, however, on the outside of the plasma membrane and is carried out by membrane-bound metalloproteases. How the intracellular domain modification communicates with the ectodomain of the substrate to allow for cleavage to occur is unknown. Here, we show that homodimerization of a cluster-of-differentiation-44 or of pro-neuregulin-1 monomers represents an essential pre-condition for their regulated ectodomain cleavage. Both substrates are associated with their respective metalloproteases under both basal or cleavage-stimulated conditions. These interactions only turn productive by specific intracellular signal-induced intracellular domain modifications of the substrates, which in turn regulate metalloprotease access to the substrates' ectodomain and cleavage. We propose that substrate intracellular domain modification induces a relative rotation or other positional change of the dimerization partners that allow metalloprotease cleavage in the extracellular space. Our findings fill an important gap in understanding substrate-specific inside-out signal transfer along cleaved transmembrane proteins and suggest that substrate dimerization (homo- or possibly heterodimerization) might represent a general principle in ectodomain shedding.


Asunto(s)
Receptores de Hialuranos/metabolismo , Neurregulina-1/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Bovinos , Línea Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Receptores de Hialuranos/química , Receptores de Hialuranos/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Células 3T3 NIH , Neurregulina-1/química , Neurregulina-1/genética , Multimerización de Proteína , Estructura Terciaria de Proteína , Proteolisis , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Especificidad por Sustrato
17.
Stem Cells ; 33(6): 1892-901, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25753288

RESUMEN

The immunomodulatory capacity of mesenchymal stem cells (MSCs) is critical for their use in therapeutic applications. MSC response to specific inflammatory cues allows them to switch between a proinflammatory (MSC1) or anti-inflammatory (MSC2) phenotype. Regulatory mechanisms controlling this switch remain to be defined. One characteristic feature of MSC2 is their ability to respond to IFNγ with induction of indoleamine 2,3-dioxygenase (IDO), representing the key immunoregulatory molecule released by human MSC. Here, we show that STAT1 and PI3Kα pathways interplay regulates IFNγ-induced IDO production in MSC. Chemical phosphoinositide 3-kinase (PI3K) pan-inhibition, PI3Kα-specific inhibition or shRNA knockdown diminished IFNγ-induced IDO production. This effect involved PI3Kα-mediated upregulation of STAT1 protein levels and phosphorylation at Ser727. Overexpression of STAT1 or of a constitutively active PI3Kα mutant failed to induce basal IDO production, but shifted MSC into an MSC2-like phenotype by strongly enhancing IDO production in response to IFNγ as compared to controls. STAT1 overexpression strongly enhanced MSC-mediated T-cell suppression. The same effect could be induced using short-term pretreatment of MSC with a chemical inhibitor of the counter player of PI3K, phosphatase and tensin homolog. Finally, downregulation of STAT1 abrogated the immunosuppressive capacity of MSC. Our results for the first time identify critical upstream signals for the induced production of IDO in MSCs that could be manipulated therapeutically to enhance their immunosuppressive phenotype.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Transcripción STAT1/metabolismo , Fosfatidilinositol 3-Quinasa Clase I , Regulación hacia Abajo , Humanos , Interferón gamma/metabolismo , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba
18.
Mol Cancer Res ; 13(5): 879-90, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25652588

RESUMEN

UNLABELLED: Ectodomain cleavage (shedding) of transmembrane proteins by metalloproteases (MMP) generates numerous essential signaling molecules, but its regulation is not totally understood. CD44, a cleaved transmembrane glycoprotein, exerts both antiproliferative or tumor-promoting functions, but whether proteolysis is required for this is not certain. CD44-mediated contact inhibition and cellular proliferation are regulated by counteracting CD44 C-terminal interacting proteins, the tumor suppressor protein merlin (NF2) and ERM proteins (ezrin, radixin, moesin). We show here that activation or overexpression of constitutively active merlin or downregulation of ERMs inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced [as well as serum, hepatocyte growth factor (HGF), or platelet-derived growth factor (PDGF)] CD44 cleavage by the metalloprotease ADAM10, whereas overexpressed ERM proteins promoted cleavage. Merlin- and ERM-modulated Ras or Rac activity was not required for this function. However, latrunculin (an actin-disrupting toxin) or an ezrin mutant which is unable to link CD44 to actin, inhibited CD44 cleavage, identifying a cytoskeletal C-terminal link as essential for induced CD44 cleavage. Cellular migration, an important tumor property, depended on CD44 and its cleavage and was inhibited by merlin. These data reveal a novel function of merlin and suggest that CD44 cleavage products play a tumor-promoting role. Neuregulin, an EGF ligand released by ADAM17 from its pro-form NRG1, is predominantly involved in regulating cellular differentiation. In contrast to CD44, release of neuregulin from its pro-form was not regulated by merlin or ERM proteins. Disruption of the actin cytoskeleton however, also inhibited NRG1 cleavage. This current study presents one of the first examples of substrate-selective cleavage regulation. IMPLICATIONS: Investigating transmembrane protein cleavage and their regulatory pathways have provided new molecular insight into their important role in cancer formation and possible treatment.


Asunto(s)
Movimiento Celular/fisiología , Receptores de Hialuranos/metabolismo , Neurofibromina 2/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Fibroblastos , Humanos , Ratones , Ratones Transgénicos , Células 3T3 NIH , Células Tumorales Cultivadas
19.
Oncotarget ; 5(19): 9281-94, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25238142

RESUMEN

In head and neck squamous cell cancer (HNSCC), four intrinsic subtypes (or groups) have been identified, and each one possesses a unique biology that will require specific treatment strategies. We previously reported that mesenchymal (group 2) tumors exhibit reduced levels of Trop2 expression. In this study, we investigated the functional role of Trop2 in HNSCC and find that loss results in autocrine activation of the EGFR family member ErbB3 via neuregulin-1. Trop2 localizes to both the cell surface and cytosol of HNSCC cells and forms a complex with neuregulin-1, which is predominantly cytosolic. Inactivation of Trop2 increases the concentration of neuregulin-1 at the cell surface where it is cleaved to activate ErbB3. In primary HNSCC, detection of ErbB3 activation was limited to Trop2 negative tumors. An analysis of the Cancer Genome Atlas (TCGA) HNSCC dataset confirms enrichment for ErbB3 activity in mesenchymal tumors. Notably, Trop2 loss triggers sensitivity to anti-ErbB3 antibodies, which results in reduced proliferation and tumorigenic growth of Trop2 negative HNSCC cancer cells. These results uncover a molecular mechanism by which tumor cells control the amount of cell-surface neuregulin-1 available for cleavage and ErbB3 activation. Moreover, we demonstrate that Trop2 is a potential surrogate biomarker to identify tumors with ErbB3 activation and may therefore respond to anti-ErbB3 therapeutics.


Asunto(s)
Antígenos de Neoplasias/genética , Carcinoma de Células Escamosas/genética , Moléculas de Adhesión Celular/genética , Neoplasias de Cabeza y Cuello/genética , Neurregulina-1/metabolismo , Receptor ErbB-3/metabolismo , Animales , Anticuerpos Antineoplásicos/inmunología , Carcinoma de Células Escamosas/clasificación , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Activación Enzimática/genética , Femenino , Neoplasias de Cabeza y Cuello/clasificación , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Interferencia de ARN , ARN Interferente Pequeño , Receptor ErbB-3/inmunología , Transducción de Señal/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Trasplante Heterólogo
20.
Proc Natl Acad Sci U S A ; 110(24): 9776-81, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23720309

RESUMEN

Ectodomain cleavage of cell-surface proteins by A disintegrin and metalloproteinases (ADAMs) is highly regulated, and its dysregulation has been linked to many diseases. ADAM10 and ADAM17 cleave most disease-relevant substrates. Broad-spectrum metalloprotease inhibitors have failed clinically, and targeting the cleavage of a specific substrate has remained impossible. It is therefore necessary to identify signaling intermediates that determine substrate specificity of cleavage. We show here that phorbol ester or angiotensin II-induced proteolytic release of EGF family members may not require a significant increase in ADAM17 protease activity. Rather, inducers activate a signaling pathway using PKC-α and the PKC-regulated protein phosphatase 1 inhibitor 14D that is required for ADAM17 cleavage of TGF-α, heparin-binding EGF, and amphiregulin. A second pathway involving PKC-δ is required for neuregulin (NRG) cleavage, and, indeed, PKC-δ phosphorylation of serine 286 in the NRG cytosolic domain is essential for induced NRG cleavage. Thus, signaling-mediated substrate selection is clearly distinct from regulation of enzyme activity, an important mechanism that offers itself for application in disease.


Asunto(s)
Proteínas ADAM/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador alfa/metabolismo , Proteínas ADAM/genética , Proteína ADAM17 , Anfirregulina , Angiotensina II/farmacología , Western Blotting , Línea Celular Tumoral , Familia de Proteínas EGF , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isoenzimas/metabolismo , Células Jurkat , Ligandos , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas/metabolismo , Proteolisis/efectos de los fármacos , Interferencia de ARN , Serina/genética , Serina/metabolismo , Especificidad por Sustrato , Acetato de Tetradecanoilforbol/farmacología , Factor de Crecimiento Transformador alfa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...