Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int Immunol ; 33(8): 435-446, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34235533

ABSTRACT

Regulatory T (Treg) cells, which are essential for maintaining self-tolerance, inhibit anti-tumor immunity, consequently hindering protective cancer immunosurveillance, and hampering effective anti-tumor immune responses in tumor-bearing hosts. Here, we show that depletion of Treg cells via targeting glycoprotein A repetitions predominant (GARP) induces effective anti-tumor immune responses. GARP was specifically expressed by highly suppressive Treg cells in the tumor microenvironment (TME) of multiple cancer types in humans. In the periphery, GARP was selectively induced in Treg cells, but not in effector T cells, by polyclonal stimulation. DS-1055a, a novel afucosylated anti-human GARP monoclonal antibody, efficiently depleted GARP+ Treg cells, leading to the activation of effector T cells. Moreover, DS-1055a decreased FoxP3+CD4+ T cells in the TME and exhibited remarkable anti-tumor activity in humanized mice bearing HT-29 tumors. We propose that DS-1055a is a new Treg-cell-targeted cancer immunotherapy agent with augmentation of anti-tumor immunity.


Subject(s)
Antibodies, Monoclonal/immunology , Membrane Proteins/immunology , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Humans , Immune Tolerance/immunology , Immunity/immunology , Immunotherapy/methods , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Tumor Microenvironment/immunology
2.
Nat Med ; 13(9): 1060-9, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17694067

ABSTRACT

Transformation and cancer growth are regulated by the coordinate actions of oncogenes and tumor suppressors. Here, we show that the novel E3 ubiquitin ligase HACE1 is frequently downregulated in human tumors and maps to a region of chromosome 6q21 implicated in multiple human cancers. Genetic inactivation of HACE1 in mice results in the development of spontaneous, late-onset cancer. A second hit from either environmental triggers or genetic heterozygosity of another tumor suppressor, p53, markedly increased tumor incidence in a Hace1-deficient background. Re-expression of HACE1 in human tumor cells directly abrogates in vitro and in vivo tumor growth, whereas downregulation of HACE1 via siRNA allows non-tumorigenic human cells to form tumors in vivo. Mechanistically, the tumor-suppressor function of HACE1 is dependent on its E3 ligase activity and HACE1 controls adhesion-dependent growth and cell cycle progression during cell stress through degradation of cyclin D1. Thus, HACE1 is a candidate chromosome 6q21 tumor-suppressor gene involved in multiple cancers.


Subject(s)
Chromosomes, Human, Pair 6 , Genes, Tumor Suppressor , Kidney Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics , Wilms Tumor/genetics , Animals , Ankyrin Repeat/genetics , Chromosome Mapping , DNA Methylation , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Mice , Mice, Mutant Strains , RNA, Messenger/genetics
3.
PLoS One ; 19(6): e0304985, 2024.
Article in English | MEDLINE | ID: mdl-38843278

ABSTRACT

Signal regulatory protein alpha (SIRPα) is an immune inhibitory receptor on myeloid cells including macrophages and dendritic cells, which binds to CD47, a ubiquitous self-associated molecule. SIRPα-CD47 interaction is exploited by cancer cells to suppress anti-tumor activity of myeloid cells, therefore emerging as a novel immune checkpoint for cancer immunotherapy. In blood cancer, several SIRPα-CD47 blockers have shown encouraging monotherapy activity. However, the anti-tumor activity of SIRPα-CD47 blockers in solid tumors seems limited, suggesting the need for combination therapies to fully exploit the myeloid immune checkpoint in solid tumors. Here we tested whether combination of SIRPα-CD47 blocker with antibody-drug conjugate bearing a topoisomerase I inhibitor DXd (DXd-ADC) would enhance anti-tumor activity in solid tumors. To this end, DS-1103a, a newly developed anti-human SIRPα antibody (Ab), was assessed for the potential combination benefit with datopotamab deruxtecan (Dato-DXd) and trastuzumab deruxtecan (T-DXd), DXd-ADCs targeting human trophoblast cell-surface antigen 2 and human epidermal growth factor receptor 2, respectively. DS-1103a inhibited SIRPα-CD47 interaction and enhanced antibody-dependent cellular phagocytosis of Dato-DXd and T-DXd against human cancer cells. In a whole cancer cell vaccination model, vaccination with DXd-treated cancer cells led to activation of tumor-specific T cells when combined with an anti-mouse SIRPα (anti-mSIRPα) Ab, implying the benefit of combining DXd-ADCs with anti-SIRPα Ab on anti-tumor immunity. Furthermore, in syngeneic mouse models, both Dato-DXd and T-DXd combination with anti-mSIRPα Ab showed stronger anti-tumor activity over the monotherapies. Taken together, this study provides a preclinical rationale of novel therapies for solid tumors combining SIRPα-CD47 blockers with DXd-ADCs.


Subject(s)
Antigens, Differentiation , CD47 Antigen , Immunoconjugates , Receptors, Immunologic , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , Animals , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Receptors, Immunologic/immunology , Humans , Mice , Immunoconjugates/pharmacology , Antigens, Differentiation/immunology , Cell Line, Tumor , Female , Trastuzumab/pharmacology , Topoisomerase I Inhibitors/pharmacology , Immunotherapy/methods , Mice, Inbred BALB C
4.
J Exp Med ; 204(4): 879-91, 2007 Apr 16.
Article in English | MEDLINE | ID: mdl-17403934

ABSTRACT

The concept of tumor surveillance implies that specific and nonspecific components of the immune system eliminate tumors in the early phase of malignancy. Understanding the biochemical mechanisms of tumor immunosurveillance is of paramount significance because it might allow one to specifically modulate spontaneous antitumor activity. We report that inactivation of the E3 ligase Casitas B cell lymphoma-b (Cbl-b) confers spontaneous in vivo rejection of tumor cells that express human papilloma virus antigens. Moreover, cbl-b(-/-) mice develop significantly fewer ultraviolet B (UVB)-induced skin malignancies and reject UVB-induced skin tumors. CD8(+) T cells were identified as key players in the spontaneous tumor rejection response. Loss of Cbl-b not only enhances antitumor reactivity of CD8(+) T cells but also occurs in the absence of CD4(+) T cells. Mechanistically, cbl-b(-/-) CD8(+) T cells are resistant to T regulatory cell-mediated suppression and exhibit enhanced activation and rapid tumor infiltration. Importantly, therapeutic transfer of naive cbl-b(-/-) CD8(+) T cells is sufficient to mediate rejection of established tumors. Even up to 1 yr after the first encounter with the tumor cells, cbl-b(-/-) mice carry an "anticancer memory." These data identify Cbl-b as a key signaling molecule that controls spontaneous antitumor activity of cytotoxic T cells in different cancer models. Inhibition of Cbl-b is a novel approach to stimulate long-lasting immunity against cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , CD8-Positive T-Lymphocytes/metabolism , Neoplasms/immunology , Proto-Oncogene Proteins c-cbl/deficiency , Proto-Oncogene Proteins c-cbl/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Female , Immunologic Memory , Interleukin-2 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Neoplasms/genetics , Neoplasms/pathology , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/immunology , Survival Rate , Ultraviolet Rays/adverse effects
5.
Nat Med ; 11(4): 394-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15750601

ABSTRACT

Morphogenesis and remodeling of bone involve synthesis of bone matrix by osteoblasts and coordinate resorption of bone by osteoclasts. Defective bone remodeling caused by altered osteoclast activity underlies a multitude of osteopenic disorders. Receptor activator of NF-kappaB (RANK) and its ligand RANKL have been identified as essential factors involved in osteoclast development and bone remodeling, but their mechanism and interacting factors have not been fully characterized. Here we report that the molecular adapter Grb-2-associated binder-2 (Gab2) associates with RANK and mediates RANK-induced activation of NF-kappaB, Akt and Jnk. Inactivation of the gene encoding Gab2 in mice results in osteopetrosis and decreased bone resorption as a result of defective osteoclast differentiation. We also show that Gab2 has a crucial role in the differentiation of human progenitor cells into osteoclasts. We have thus identified a new, key regulatory scaffold molecule, Gab2, that controls select RANK signaling pathways and is essential for osteoclastogenesis and bone homeostasis.


Subject(s)
Carrier Proteins/physiology , Membrane Glycoproteins/physiology , Osteoclasts/physiology , Osteogenesis , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Bone Remodeling , Bone Resorption , Cell Differentiation , Cells, Cultured , Humans , Mice , Mice, Mutant Strains , Mutation , Osteoblasts/physiology , Osteopetrosis/genetics , Phosphoproteins/genetics , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Signal Transduction
6.
Nat Med ; 11(8): 875-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16007097

ABSTRACT

During several months of 2003, a newly identified illness termed severe acute respiratory syndrome (SARS) spread rapidly through the world. A new coronavirus (SARS-CoV) was identified as the SARS pathogen, which triggered severe pneumonia and acute, often lethal, lung failure. Moreover, among infected individuals influenza such as the Spanish flu and the emergence of new respiratory disease viruses have caused high lethality resulting from acute lung failure. In cell lines, angiotensin-converting enzyme 2 (ACE2) has been identified as a potential SARS-CoV receptor. The high lethality of SARS-CoV infections, its enormous economic and social impact, fears of renewed outbreaks as well as the potential misuse of such viruses as biologic weapons make it paramount to understand the pathogenesis of SARS-CoV. Here we provide the first genetic proof that ACE2 is a crucial SARS-CoV receptor in vivo. SARS-CoV infections and the Spike protein of the SARS-CoV reduce ACE2 expression. Notably, injection of SARS-CoV Spike into mice worsens acute lung failure in vivo that can be attenuated by blocking the renin-angiotensin pathway. These results provide a molecular explanation why SARS-CoV infections cause severe and often lethal lung failure and suggest a rational therapy for SARS and possibly other respiratory disease viruses.


Subject(s)
Carboxypeptidases/metabolism , Lung Diseases/enzymology , Membrane Glycoproteins/metabolism , Severe Acute Respiratory Syndrome/enzymology , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/metabolism , Analysis of Variance , Angiotensin-Converting Enzyme 2 , Animals , Immunohistochemistry , Lung Diseases/etiology , Lung Diseases/pathology , Membrane Glycoproteins/genetics , Mice , Peptidyl-Dipeptidase A , Severe acute respiratory syndrome-related coronavirus/genetics , Severe Acute Respiratory Syndrome/complications , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics
7.
Nature ; 442(7101): 457-60, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16871217

ABSTRACT

Wound healing is essential for maintaining the integrity of multicellular organisms. In every species studied, disruption of an epithelial layer instantaneously generates endogenous electric fields, which have been proposed to be important in wound healing. The identity of signalling pathways that guide both cell migration to electric cues and electric-field-induced wound healing have not been elucidated at a genetic level. Here we show that electric fields, of a strength equal to those detected endogenously, direct cell migration during wound healing as a prime directional cue. Manipulation of endogenous wound electric fields affects wound healing in vivo. Electric stimulation triggers activation of Src and inositol-phospholipid signalling, which polarizes in the direction of cell migration. Notably, genetic disruption of phosphatidylinositol-3-OH kinase-gamma (PI(3)Kgamma) decreases electric-field-induced signalling and abolishes directed movements of healing epithelium in response to electric signals. Deletion of the tumour suppressor phosphatase and tensin homolog (PTEN) enhances signalling and electrotactic responses. These data identify genes essential for electrical-signal-induced wound healing and show that PI(3)Kgamma and PTEN control electrotaxis.


Subject(s)
PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Wound Healing , Animals , Cell Movement , Class Ib Phosphatidylinositol 3-Kinase , Dictyostelium , Electric Stimulation , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction , Wound Healing/genetics
8.
Nature ; 440(7084): 692-6, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16572175

ABSTRACT

Bone metastases are a frequent complication of many cancers that result in severe disease burden and pain. Since the late nineteenth century, it has been thought that the microenvironment of the local host tissue actively participates in the propensity of certain cancers to metastasize to specific organs, and that bone provides an especially fertile 'soil'. In the case of breast cancers, the local chemokine milieu is now emerging as an explanation for why these tumours preferentially metastasize to certain organs. However, as the inhibition of chemokine receptors in vivo only partially blocks metastatic behaviour, other factors must exist that regulate the preferential metastasis of breast cancer cells. Here we show that the cytokine RANKL (receptor activator of NF-kappaB ligand) triggers migration of human epithelial cancer cells and melanoma cells that express the receptor RANK. RANK is expressed on cancer cell lines and breast cancer cells in patients. In a mouse model of melanoma metastasis, in vivo neutralization of RANKL by osteoprotegerin results in complete protection from paralysis and a marked reduction in tumour burden in bones but not in other organs. Our data show that local differentiation factors such as RANKL have an important role in cell migration and the tissue-specific metastatic behaviour of cancer cells.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Carrier Proteins/metabolism , Cell Movement , Membrane Glycoproteins/metabolism , Neoplasm Metastasis/pathology , Animals , Bone Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Cell Death , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Epithelial Cells/pathology , Female , Humans , Melanoma/metabolism , Melanoma/pathology , Membrane Glycoproteins/genetics , Mice , Organ Specificity , Paralysis , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Signal Transduction
9.
J Exp Med ; 201(6): 859-70, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15767368

ABSTRACT

The membrane phospholipid phosphatidylinositol 4, 5-bisphosphate [PI(4,5)P(2)] is a critical signal transducer in eukaryotic cells. However, the physiological roles of the type I phosphatidylinositol phosphate kinases (PIPKIs) that synthesize PI(4,5)P(2) are largely unknown. Here, we show that the alpha isozyme of PIPKI (PIPKIalpha) negatively regulates mast cell functions and anaphylactic responses. In vitro, PIPKIalpha-deficient mast cells exhibited increased degranulation and cytokine production after Fcepsilon receptor-I cross-linking. In vivo, PIPKIalpha(-/-) mice displayed enhanced passive cutaneous and systemic anaphylaxis. Filamentous actin was diminished in PIPKIalpha(-/-) mast cells, and enhanced degranulation observed in the absence of PIPKIalpha was also seen in wild-type mast cells treated with latrunculin, a pharmacological inhibitor of actin polymerization. Moreover, the association of FcepsilonRI with lipid rafts and FcepsilonRI-mediated activation of signaling proteins was augmented in PIPKIalpha(-/-) mast cells. Thus, PIPKIalpha is a negative regulator of FcepsilonRI-mediated cellular responses and anaphylaxis, which functions by controlling the actin cytoskeleton and dynamics of FcepsilonRI signaling. Our results indicate that the different PIPKI isoforms might be functionally specialized.


Subject(s)
Anaphylaxis/metabolism , Calcium Signaling/physiology , Cell Degranulation/physiology , Mast Cells/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Actins/metabolism , Anaphylaxis/genetics , Animals , Calcium Signaling/genetics , Cell Degranulation/genetics , Cells, Cultured , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Minor Histocompatibility Antigens , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, IgE/metabolism , Thiazoles/pharmacology
10.
Nat Cell Biol ; 6(3): 215-26, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15039780

ABSTRACT

During the development of multicellular organisms, concerted actions of molecular signalling networks determine whether cells undergo proliferation, differentiation, death or ageing. Here we show that genetic inactivation of the stress signalling kinase, MKK7, a direct activator of JNKs in mice, results in embryonic lethality and impaired proliferation of hepatocytes. Beginning at passage 4-5, mkk7(-/-) mouse embryonic fibroblasts (MEFs) display impaired proliferation, premature senescence and G2/M cell cycle arrest. Similarly, loss of c-Jun or expression of a c-JunAA mutant in which the JNK phosphorylation sites were replaced with alanine results in a G2/M cell-cycle block. The G2/M cell-cycle kinase CDC2 was identified as a target for the MKK7-JNK-c-Jun pathway. These data show that the MKK7-JNK-c-Jun signalling pathway couples developmental and environmental cues to CDC2 expression, G2/M cell cycle progression and cellular senescence in fibroblasts.


Subject(s)
Cellular Senescence/genetics , G2 Phase/genetics , Hepatocytes/enzymology , Mitogen-Activated Protein Kinase Kinases/deficiency , Mitosis/genetics , Stress, Physiological/enzymology , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Fetus , Fibroblasts/cytology , Fibroblasts/enzymology , Genes, Lethal/genetics , Hepatocytes/cytology , JNK Mitogen-Activated Protein Kinases , Liver/abnormalities , Liver/pathology , MAP Kinase Kinase 7 , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Phosphorylation , Proto-Oncogene Proteins c-jun/deficiency , Proto-Oncogene Proteins c-jun/genetics , Stress, Physiological/genetics
11.
Nature ; 436(7047): 112-6, 2005 Jul 07.
Article in English | MEDLINE | ID: mdl-16001071

ABSTRACT

Acute respiratory distress syndrome (ARDS), the most severe form of acute lung injury, is a devastating clinical syndrome with a high mortality rate (30-60%) (refs 1-3). Predisposing factors for ARDS are diverse and include sepsis, aspiration, pneumonias and infections with the severe acute respiratory syndrome (SARS) coronavirus. At present, there are no effective drugs for improving the clinical outcome of ARDS. Angiotensin-converting enzyme (ACE) and ACE2 are homologues with different key functions in the renin-angiotensin system. ACE cleaves angiotensin I to generate angiotensin II, whereas ACE2 inactivates angiotensin II and is a negative regulator of the system. ACE2 has also recently been identified as a potential SARS virus receptor and is expressed in lungs. Here we report that ACE2 and the angiotensin II type 2 receptor (AT2) protect mice from severe acute lung injury induced by acid aspiration or sepsis. However, other components of the renin-angiotensin system, including ACE, angiotensin II and the angiotensin II type 1a receptor (AT1a), promote disease pathogenesis, induce lung oedemas and impair lung function. We show that mice deficient for Ace show markedly improved disease, and also that recombinant ACE2 can protect mice from severe acute lung injury. Our data identify a critical function for ACE2 in acute lung injury, pointing to a possible therapy for a syndrome affecting millions of people worldwide every year.


Subject(s)
Carboxypeptidases/metabolism , Lung Diseases/enzymology , Lung Diseases/pathology , Acids/pharmacology , Acute Disease , Analysis of Variance , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Capillary Permeability , Carboxypeptidases/deficiency , Carboxypeptidases/genetics , Disease Models, Animal , Gene Deletion , Humans , Lung/pathology , Lung/physiopathology , Lung Diseases/genetics , Lung Diseases/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Oxygen/blood , Peptidyl-Dipeptidase A , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/pathology , Sepsis/enzymology , Sepsis/genetics , Sepsis/pathology , Transgenes/genetics
12.
Sci Rep ; 11(1): 341, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431933

ABSTRACT

Single-cell level analysis is powerful tool to assess the heterogeneity of cellular components in tumor microenvironments (TME). In this study, we investigated immune-profiles using the single-cell analyses of endoscopically- or surgically-resected tumors, and peripheral blood mononuclear cells from gastric cancer patients. Furthermore, we technically characterized two distinct platforms of the single-cell analysis; RNA-seq-based analysis (scRNA-seq), and mass cytometry-based analysis (CyTOF), both of which are broadly embraced technologies. Our study revealed that the scRNA-seq analysis could cover a broader range of immune cells of TME in the biopsy-resected small samples of tumors, detecting even small subgroups of B cells or Treg cells in the tumors, although CyTOF could distinguish the specific populations in more depth. These findings demonstrate that scRNA-seq analysis is a highly-feasible platform for elucidating the complexity of TME in small biopsy tumors, which would provide a novel strategies to overcome a therapeutic difficulties against cancer heterogeneity in TME.


Subject(s)
Single-Cell Analysis , Stomach Neoplasms/pathology , Tumor Microenvironment , Adult , Biopsy , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , RNA-Seq , Stomach Neoplasms/genetics
13.
J Exp Med ; 200(9): 1167-77, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15520247

ABSTRACT

Carma1 (also known as caspase recruitment domain [CARD]11, Bimp3) is a CARD-containing membrane-associated guanylate kinase family protein that plays an essential role in antigen receptor-induced nuclear factor kappaB activation. We investigated the role of Carma1 in the assembly of signaling molecules at the immune synapse using a peptide-specific system. We report that Carma1 is essential for peptide-induced interleukin 2 and interferon gamma production, but dispensable for proliferation in T cells. Recruitment and distribution of T cell receptor, lymphocyte function associated 1, lipid rafts, and protein kinase C (PKC)theta; to central and peripheral immune synapse regions occur normally in Carma1-/- T cells. Carma1 controls entry of IkappaB kinase (IKK) into lipid raft aggregates and the central region of the immune synapse, as well as activation of IKK downstream of PKC. Our data provide the first genetic evidence on a new class of molecular scaffold that controls entry of defined signaling components, IKK, into the central supramolecular activation cluster at T cell-antigen-presenting cell interfaces without having any apparent effect on the overall organization and formation of immune synapses.


Subject(s)
Antigen Presentation/immunology , Nucleoside-Phosphate Kinase/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Adhesion/immunology , Cell Fractionation , Flow Cytometry , Fluorescent Antibody Technique , Guanylate Kinases , I-kappa B Kinase , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Transgenic , Microscopy, Confocal , Nucleoside-Phosphate Kinase/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism
14.
FASEB Bioadv ; 2(8): 478-488, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32821879

ABSTRACT

ORAI1 constitutes the pore-forming subunit of the calcium release-activated calcium (CRAC) channel, which is responsible for store-operated calcium entry into lymphocytes. It is known that ORAI1 is essential for the activation of T cells and mast cells and is considered to be a potent therapeutic target for autoimmune and allergic diseases. Here, we obtained a new humanized antibody, DS-2741a, that inhibits ORAI1 function. DS-2741a bound to human-ORAI1 with high affinity and without cross-reactivity to rodent Orai1. DS-2741a demonstrated suppression of CRAC-mediated human and mouse T-cell activation and mast cell degranulation in human ORAI1 knock-in mice. Furthermore, DS-2741a ameliorated house dust mite antigen-induced dermatitis in the human ORAI1 knock-in mouse. Taken together, DS-2741a inhibited T-cell and mast cell functions, thus improving skin inflammation in animal models of atopic dermatitis and reinforcing the need for investigation of DS-2741a for the treatment of allergic diseases in a clinical setting.

15.
PLoS One ; 14(10): e0222280, 2019.
Article in English | MEDLINE | ID: mdl-31574081

ABSTRACT

[Fam-] trastuzumab deruxtecan (DS-8201a) is a HER2 (ERBB2)-targeting antibody-drug conjugate, composed of a HER2-targeting antibody and a topoisomerase I inhibitor, exatecan derivative, that has antitumor effects in preclinical xenograft models and clinical trials. Recently, [fam-] trastuzumab deruxtecan was reported to enhance antitumor immunity and was beneficial in combination with an anti-PD-1 antibody in a mouse model. In this study, the antitumor effect of [fam-] trastuzumab deruxtecan in combination with an anti-CTLA-4 antibody was evaluated. [Fam-] trastuzumab deruxtecan monotherapy had antitumor activity in an immunocompetent mouse model with EMT6 human HER2-expressing mouse breast cancer cells (EMT6-hHER2). [Fam-] trastuzumab deruxtecan in combination with the anti-CTLA-4 antibody induced more potent antitumor activity than that by monotherapy with either agent. The combination therapy increased tumor-infiltrating CD4+ and CD8+ T cells in vivo. Mechanistically, cured mice with treatment of [fam-] trastuzumab deruxtecan and an anti-CTLA-4 antibody completely rejected EMT6-mock cells similar to EMT6-hHER2 cells, and splenocytes from the cured mice responded to both EMT6-hHER2 and EMT6-mock cells as measured by interferon-gamma release. Taken together, these results indicate that antitumor immunity is induced by [fam-] trastuzumab deruxtecan and is facilitated in combination with anti-CTLA-4 antibody.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/drug therapy , CTLA-4 Antigen/immunology , Camptothecin/analogs & derivatives , Immunity, Innate/drug effects , Immunoconjugates/pharmacology , Animals , Antibodies/drug effects , Antibodies/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Mice , Receptor, ErbB-2/genetics , Trastuzumab
16.
Mol Cell Biol ; 25(23): 10261-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16287843

ABSTRACT

Cardiac and skeletal muscle critically depend on mitochondrial energy metabolism for their normal function. Recently, we showed that apoptosis-inducing factor (AIF), a mitochondrial protein implicated in programmed cell death, plays a role in mitochondrial respiration. However, the in vivo consequences of AIF-regulated mitochondrial respiration resulting from a loss-of-function mutation in Aif are not known. Here, we report tissue-specific deletion of Aif in the mouse. Mice in which Aif has been inactivated specifically in cardiac and skeletal muscle exhibit impaired activity and protein expression of respiratory chain complex I. Mutant animals develop severe dilated cardiomyopathy, heart failure, and skeletal muscle atrophy accompanied by lactic acidemia consistent with defects in the mitochondrial respiratory chain. Isolated hearts from mutant animals exhibit poor contractile performance in response to a respiratory chain-dependent energy substrate, but not in response to glucose, supporting the notion that impaired heart function in mutant animals results from defective mitochondrial energy metabolism. These data provide genetic proof that the previously defined cell death promoter AIF has a second essential function in mitochondrial respiration and aerobic energy metabolism required for normal heart function and skeletal muscle homeostasis.


Subject(s)
Apoptosis Inducing Factor/deficiency , Apoptosis Inducing Factor/metabolism , Cardiomyopathy, Dilated/pathology , Mitochondria/metabolism , Mitochondria/pathology , Muscular Atrophy/pathology , Animals , Apoptosis Inducing Factor/genetics , Biomarkers , Cardiomyopathy, Dilated/embryology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Glucose/metabolism , Mice , Mice, Transgenic , Muscular Atrophy/embryology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Mutation/genetics , Oxidative Stress
17.
Mol Cancer Ther ; 17(7): 1494-1503, 2018 07.
Article in English | MEDLINE | ID: mdl-29703841

ABSTRACT

Trastuzumab deruxtecan (DS-8201a), a HER2-targeting antibody-drug conjugate with a topoisomerase I inhibitor exatecan derivative (DX-8951 derivative, DXd), has been reported to exert potent antitumor effects in xenograft mouse models and clinical trials. In this study, the immune system-activating ability of DS-8201a was assessed. DS-8201a significantly suppressed tumor growth in an immunocompetent mouse model with human HER2-expressing CT26.WT (CT26.WT-hHER2) cells. Cured immunocompetent mice rejected not only rechallenged CT26.WT-hHER2 cells, but also CT26.WT-mock cells. Splenocytes from the cured mice responded to both CT26.WT-hHER2 and CT26.WT-mock cells. Further analyses revealed that DXd upregulated CD86 expression on bone marrow-derived dendritic cells (DC) in vitro and that DS-8201a increased tumor-infiltrating DCs and upregulated their CD86 expression in vivo DS-8201a also increased tumor-infiltrating CD8+ T cells and enhanced PD-L1 and MHC class I expression on tumor cells. Furthermore, combination therapy with DS-8201a and anti-PD-1 antibody was more effective than either monotherapy. In conclusion, DS-8201a enhanced antitumor immunity, as evidenced by the increased expression of DC markers, augmented expression of MHC class I in tumor cells, and rejection of rechallenged tumor cells by adaptive immune cells, suggesting that DS-8201a enhanced tumor recognition by T cells. Furthermore, DS-8201a treatment benefited from combination with anti-PD-1 antibody, possibly due to increased T-cell activity and upregulated PD-L1 expression induced by DS-8201a. Mol Cancer Ther; 17(7); 1494-503. ©2018 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Immunoconjugates/administration & dosage , Trastuzumab/administration & dosage , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Camptothecin/administration & dosage , Camptothecin/chemistry , Camptothecin/immunology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/immunology , Trastuzumab/chemistry , Trastuzumab/immunology , Xenograft Model Antitumor Assays
19.
Trends Mol Med ; 12(1): 17-25, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16356770

ABSTRACT

Hundreds of millions of people worldwide are affected by bone-related diseases, such as osteoporosis and rheumatoid arthritis. Understanding the molecular mechanisms of bone metabolism is crucial for developing novel drugs for treating such diseases. In particular, genetic experiments showing that the receptor activator of NF-kappaB (RANK), its ligand RANKL, and the decoy receptor OPG are essential, central regulators of osteoclast development and osteoclast function were significant turning points in our understanding of bone diseases. RANKL-RANK signaling activates a variety of downstream signaling pathways required for osteoclast development. Moreover, molecular cross-talk between RANKL-RANK and other ligand-receptor systems fine-tunes bone homeostasis in normal physiology and disease. Designing novel drugs that target RANKL-RANK and their signaling pathways in osteoclasts could potentially revolutionize the treatment of many diseases associated with bone loss such as arthritis, tooth loss, cancer metastases or osteoporosis.


Subject(s)
Bone Diseases/metabolism , Bone Resorption/metabolism , Carrier Proteins/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Diseases/etiology , Bone Diseases/therapy , Bone Resorption/etiology , Bone Resorption/therapy , Humans , MAP Kinase Signaling System , Models, Biological , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoprotegerin , Phosphatidylinositols/metabolism , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptor Cross-Talk , Signal Transduction , src-Family Kinases/metabolism
20.
Circ Res ; 91(10): 961-9, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12433842

ABSTRACT

In neonatal cardiomyocytes, activation of the G(q)-coupled alpha(1)-adrenergic receptor (alpha(1)AR) induces hypertrophy by activating mitogen-activated protein kinases, including c-Jun NH(2)-terminal kinase (JNK). Here, we show that JNK activation is essential for alpha(1)AR-induced hypertrophy, in that alpha(1)AR-induced hypertrophic responses, such as reorganization of the actin cytoskeleton and increased protein synthesis, could be blocked by expressing the JNK-binding domain of JNK-interacting protein-1, a specific inhibitor of JNK. We also identified the classes and subunits of G proteins that mediate alpha(1)AR-induced JNK activation and hypertrophic responses by generating several recombinant adenoviruses that express polypeptides capable of inhibiting the function of specific G-protein subunits. alpha(1)AR-induced JNK activation was inhibited by the expression of carboxyl terminal regions of Galpha(q), Galpha(12), and Galpha(13). JNK activation was also inhibited by the Galpha(q/11)- or Galpha(12/13)-specific regulator of G-protein signaling (RGS) domains and by C3 toxin but was not affected by treatment with pertussis toxin or by expression of the carboxyl terminal region of G protein-coupled receptor kinase 2, a polypeptide that sequesters Gbetagamma. alpha(1)AR-induced hypertrophic responses were inhibited by Galpha(q/11)- and Galpha(12/13)-specific RGS domains, C3 toxin, and the carboxyl terminal region of G protein-coupled receptor kinase 2 but not by pertussis toxin. Activation of Rho was inhibited by carboxyl terminal regions of Galpha(12) and Galpha(13) but not by Galpha(q). Our findings suggest that alpha(1)AR-induced hypertrophic responses are mediated in part by a Galpha(12/13)-Rho-JNK pathway, in part by a G(q/11)-JNK pathway that is Rho independent, and in part by a Gbetagamma pathway that is JNK independent.


Subject(s)
Adaptor Proteins, Signal Transducing , Cardiomegaly/metabolism , DNA-Binding Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Myocardium/metabolism , Receptors, Adrenergic, alpha-1/metabolism , ADP Ribose Transferases/pharmacology , Adenoviridae/genetics , Animals , Animals, Newborn , Botulinum Toxins/pharmacology , Cardiomegaly/etiology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cells, Cultured , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, G12-G13 , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/genetics , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Oxidants/pharmacology , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Pertussis Toxin/pharmacology , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transfection , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL