Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Nanotechnol ; 16(11): 1271-1280, 2021 11.
Article in English | MEDLINE | ID: mdl-34580467

ABSTRACT

Impaired type I interferons (IFNs) may cause immune deficiency in tumours. Current supplementary IFN therapy partially restores anticancer immunity but simultaneously induces immune evasion by upregulating multiple immune checkpoints. Here we create a T lymphocyte membrane-decorated epigenetic nanoinducer that is engineered with programmed cell death protein 1 (PD1), which we call OPEN, for the delivery of the IFN inducer ORY-1001. OPEN increases IFNs and blocks IFN-induced immune checkpoint upregulation. OPEN also targets tumours that express programmed cell death ligand 1 (PDL1) through PDL1/PD1 recognition and subsequently triggers the internalization of OPEN and immune checkpoint proteins. OPEN, which is loaded with ORY-1001, upregulates intratumoural IFNs and downstream major histocompatibility complex I and PDL1. The replenished PDL1 enables further ligation of OPEN, which in turn blocks PDL1. These sequential processes result in an eight- and 29-fold increase of the intratumoural densities of total and active cytotoxic T lymphocytes, respectively, and a strong inhibition of xenograft tumour growth. This T lymphocyte membrane-decorated epigenetic nanoinducer presents a generalizable platform to boost antitumour immunity.


Subject(s)
Immunotherapy , Interferon Type I/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Biomarkers, Tumor/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/genetics
2.
Adv Healthc Mater ; 10(1): e2001239, 2021 01.
Article in English | MEDLINE | ID: mdl-32935937

ABSTRACT

Supramolecular peptide hydrogel (SPH) is a class of biomaterials self-assembled from peptide-based gelators through non-covalent interactions. Among many of its biomedical applications, the potential of SPH in cancer therapy has been vastly explored in the past decade, taking advantage of its good biocompatibility, multifunctionality, and injectability. SPHs can exert localized cancer therapy and induce systemic anticancer immunity to prevent tumor recurrence, depending on the design of SPH. This review first gives a brief introduction to SPH and then outlines the major types of peptide-based gelators that have been developed so far. The methodologies to tune the physicochemical properties and biological activities are summarized. The recent advances of SPH in cancer therapy as carriers, prodrugs, or drugs are highlighted. Finally, the clinical translation potential and main challenges in this field are also discussed.


Subject(s)
Hydrogels , Neoplasms , Biocompatible Materials , Neoplasms/drug therapy , Peptides
3.
Cell Metab ; 20(2): 320-32, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24981838

ABSTRACT

Bile acid (BA) biosynthesis is tightly controlled by intrahepatic negative feedback signaling elicited by BA binding to farnesoid X receptor (FXR) and also by enterohepatic communication involving ileal BA reabsorption and FGF15/19 secretion. However, how these pathways are coordinated is poorly understood. We show here that nonreceptor tyrosine phosphatase Shp2 is a critical player that couples and regulates the intrahepatic and enterohepatic signals for repression of BA synthesis. Ablating Shp2 in hepatocytes suppressed signal relay from FGFR4, receptor for FGF15/19, and attenuated BA activation of FXR signaling, resulting in elevation of systemic BA levels and chronic hepatobiliary disorders in mice. Acting immediately downstream of FGFR4, Shp2 associates with FRS2α and promotes the receptor activation and signal relay to several pathways. These results elucidate a molecular mechanism for the control of BA homeostasis by Shp2 through the orchestration of multiple signals in hepatocytes.


Subject(s)
Bile Acids and Salts/biosynthesis , Liver/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction , Animals , Bile Ducts/injuries , Cell Line , Cholesterol 7-alpha-Hydroxylase/metabolism , Fibroblast Growth Factors/metabolism , Liver/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Up-Regulation
4.
J Biol Chem ; 288(6): 3823-30, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23269672

ABSTRACT

The Src-homology 2 (SH2) domain-containing tyrosine phosphatase Shp2 has been known to regulate various signaling pathways triggered by receptor and cytoplasmic tyrosine kinases. Here we describe a novel function of Shp2 in control of lipid metabolism by mediating degradation of fatty acid synthase (FASN). p38-phosphorylated COP1 accumulates in the cytoplasm and subsequently binds FASN through Shp2 here as an adapter, leading to FASN-Shp2-COP1 complex formation and FASN degradation mediated by ubiquitination pathway. By fasting p38 is activated and stimulates FASN protein degradation in mice. Consistently, the FASN protein levels are dramatically elevated in mouse liver and pancreas in which Shp2/Ptpn11 is selectively deleted. Thus, this study identifies a new activity for Shp2 in lipid metabolism.


Subject(s)
Fatty Acid Synthase, Type I/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , 3T3 Cells , Animals , Fatty Acid Synthase, Type I/genetics , HeLa Cells , Humans , Lipid Metabolism/physiology , Liver/enzymology , Mice , Mice, Knockout , Nuclear Proteins/genetics , Pancreas/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Ubiquitin-Protein Ligases/genetics , p38 Mitogen-Activated Protein Kinases/genetics
5.
Proc Natl Acad Sci U S A ; 110(1): E79-88, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23236157

ABSTRACT

The molecular mechanism underlying adipogenesis and the physiological functions of adipose tissue are not fully understood. We describe here a unique mouse model of severe lipodystrophy. Ablation of Ptpn11/Shp2 in adipocytes, mediated by aP2-Cre, led to premature death, lack of white fat, low blood pressure, compensatory erythrocytosis, and hepatic steatosis in Shp2(fat-/-) mice. Fat transplantation partially rescued the lifespan and blood pressure in Shp2(fat-/-) mice, and administration of leptin also restored partially the blood pressure of mutant animals with endogenous leptin deficiency. Consistently, homozygous deletion of Shp2 inhibited adipocyte differentiation from embryonic stem (ES) cells. Biochemical analyses suggest a Shp2-TAO2-p38-p300-PPARγ pathway in adipogenesis, in which Shp2 suppresses p38 activation, leading to stabilization of p300 and enhanced PPARγ expression. Inhibition of p38 restored adipocyte differentiation from Shp2(-/-) ES cells, and p38 signaling is also suppressed in obese patients and obese animals. These results illustrate an essential role of adipose tissue in mammalian survival and physiology and also suggest a common signaling mechanism involved in adipogenesis and obesity development.


Subject(s)
Adipogenesis/physiology , Disease Models, Animal , Lipodystrophy/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adipose Tissue/transplantation , Animals , Blood Pressure/drug effects , DNA Primers/genetics , E1A-Associated p300 Protein/metabolism , Gene Deletion , Leptin/administration & dosage , Leptin/deficiency , Leptin/pharmacology , Mice , Mice, Knockout , PPAR gamma/metabolism , Protein Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Genes Dev ; 26(14): 1587-601, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22759635

ABSTRACT

Hematopoietic development occurs in complex microenvironments and is influenced by key signaling events. Yet how these pathways communicate with master hematopoietic transcription factors to coordinate differentiation remains incompletely understood. The transcription factor RUNX1 plays essential roles in definitive hematopoietic stem cell (HSC) ontogeny, HSC maintenance, megakaryocyte (Mk) maturation, and lymphocyte differentiation. It is also the most frequent target of genetic alterations in human leukemia. Here, we report that RUNX1 is phosphorylated by Src family kinases (SFKs) and that this occurs on multiple tyrosine residues located within its negative regulatory DNA-binding and autoinhibitory domains. Retroviral transduction, chemical inhibitor, and genetic studies demonstrate a negative regulatory role of tyrosine phosphorylation on RUNX1 activity in Mk and CD8 T-cell differentiation. We also demonstrate that the nonreceptor tyrosine phosphatase Shp2 binds directly to RUNX1 and contributes to its dephosphorylation. Last, we show that RUNX1 tyrosine phosphorylation correlates with reduced GATA1 and enhanced SWI/SNF interactions. These findings link SFK and Shp2 signaling pathways to the regulation of RUNX1 activity in hematopoiesis via control of RUNX1 multiprotein complex assembly.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Megakaryocytes/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoiesis/physiology , Humans , Megakaryocytes/cytology , Mice , Mice, Transgenic , Phosphorylation/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , src-Family Kinases/genetics
7.
Mol Cell Biol ; 32(10): 1867-78, 2012 May.
Article in English | MEDLINE | ID: mdl-22431513

ABSTRACT

In mammals, leptin regulates food intake and energy balance mainly through the activation of LepRb in the hypothalamus, and estrogen has a leptin-like effect in the hypothalamic control of metabolism. However, it remains to be elucidated how estrogen signaling is intertwined with the leptin pathway. We show here that Shp2, a nonreceptor tyrosine phosphatase, acts to integrate leptin and estrogen signals. The expression of a dominant-active mutant (Shp2(D61A)) in forebrain neurons conferred female, but not male, transgenic mice resistance to high-fat diet (HFD)-induced obesity and liver steatosis, accompanied by improved insulin sensitivity and glucose homeostasis. Fed with either HFD or regular chow food, Shp2(D61A) female mice showed dramatically enhanced leptin sensitivity. Microinjection of Shp2(D61A)-expressing adeno-associated virus into mediobasal hypothalamus elicited a similar antiobese effect in female mice. Biochemical analyses showed a physical association of Shp2 with estrogen receptor alpha, which is necessary for the synergistic and persistent activation of Erk by leptin and estrogen. Together, these results elucidate a mechanism for the direct cross talk of leptin and estrogen signaling and offer one explanation for the propensity of postmenopausal women to develop obesity.


Subject(s)
Body Weight/genetics , Energy Metabolism/genetics , Estrogens/metabolism , Leptin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Animals , Diet, High-Fat , Estrogen Receptor alpha/metabolism , Female , Hypothalamus/metabolism , Male , Mice , Mice, Transgenic , Mutation , Neurons/metabolism , Obesity/genetics , Obesity/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptors, Leptin/metabolism , Sex Factors , Signal Transduction
8.
Cancer Cell ; 19(5): 629-39, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21575863

ABSTRACT

The human gene Ptpn11, which encodes the tyrosine phosphatase Shp2, may act as a proto-oncogene because dominantly activating mutations have been detected in several types of leukemia. Herein we report a tumor-suppressor function of Shp2. Hepatocyte-specific deletion of Shp2 promotes inflammatory signaling through the Stat3 pathway and hepatic inflammation/necrosis, resulting in regenerative hyperplasia and development of tumors in aged mice. Furthermore, Shp2 ablation dramatically enhanced diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) development, which was abolished by concurrent deletion of Shp2 and Stat3 in hepatocytes. Decreased Shp2 expression was detected in a subfraction of human HCC specimens. Thus, in contrast to the leukemogenic effect of dominant-active mutants, Ptpn11/Shp2 has a tumor-suppressor function in liver.


Subject(s)
Adenoma, Liver Cell/enzymology , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Liver/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Tumor Suppressor Proteins/metabolism , Adenoma, Liver Cell/genetics , Adenoma, Liver Cell/pathology , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cytokines/blood , Cytokines/genetics , Diethylnitrosamine , Gene Expression Regulation , Hepatitis/enzymology , Hepatitis/genetics , Hepatitis/pathology , Humans , Hyperplasia , Inflammation Mediators/blood , Interleukin-6/administration & dosage , Lipopolysaccharides/administration & dosage , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Liver Regeneration , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Protein Tyrosine Phosphatase, Non-Receptor Type 11/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Mas , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
9.
J Immunol ; 183(8): 4940-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19786542

ABSTRACT

Clustering of the high affinity IgE receptor (Fc(epsilon)RI) in mast cells leads to degranulation and production of numerous cytokines and lipid mediators that promote allergic inflammation. Initiation of FFc(epsilon)RI signaling involves rapid tyrosine phosphorylation of Fc(epsilon)RI and membrane-localized adaptor proteins that recruit additional SH2 domain-containing proteins that dynamically regulate downstream signaling. SH2 domain-containing phosphatase-2 (SHP2) is a protein-tyrosine phosphatase implicated in Fc(epsilon)RI signaling, but whose function is not well defined. In this study, using a mouse model allowing temporal shp2 inactivation in bone marrow-derived mast cells (BMMCs), we provide insights into SHP2 functions in the Fc(epsilon)RI pathway. Although no overt defects in Fc(epsilon)RI-induced tyrosine phosphorylation were observed in SHP2 knock-out (KO) BMMCs, several proteins including Lyn and Syk kinases displayed extended phosphorylation kinetics compared with wild-type BMMCs. SHP2 was dispensable for Fc(epsilon)RI-induced degranulation of BMMCs, but was required for maximal activation of Erk and Jnk mitogen-activated protein kinases. SHP2 KO BMMCs displayed several phenotypes associated with reduced Fyn activity, including elevated phosphorylation of the inhibitory pY531 site in Fyn, impaired signaling to Grb2-associated binder 2, Akt/PKB, and IkappaB kinase, and decreased TNF-alpha release compared with control cells. This is likely due to elevated Lyn activity in SHP2 KO BMMCs, and the ability of Lyn to antagonize Fyn activity. Overall, our study identifies SHP2 as a positive effector of Fc(epsilon)RI-induced activation of Fyn/Grb2-associated binder 2/Akt and Ras/Erk pathways leading to TNF-alpha release from mast cells.


Subject(s)
Bone Marrow Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/immunology , Mast Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins c-fyn/metabolism , Receptors, IgE/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Bone Marrow Cells/immunology , Cell Degranulation/genetics , Cell Degranulation/immunology , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Knockdown Techniques , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-fyn/physiology , src Homology Domains/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...