Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Regen Med ; 19(3): 119-133, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38449425

ABSTRACT

Aim: This study aimed to evaluate the trends in organoid culture research within the field of regenerative medicine from 2002 to 2022. Methods: The worldwide distribution of organoid research in regenerative medicine articles indexed in the Scopus database was analyzed. Result: A total of 840 documents were analyzed, averaging 42 publications annually. The USA (n = 296) led in publications, followed by China (n = 127), Japan (n = 91) and the UK (n = 75). Since 2011, research has surged, particularly in China, which emerged as a prominent center. Conclusion: The findings highlight significant growth in organoid research, promising future organ transplantation. Research trends integrate tissue engineering, gene modification and induced pluripotent stem cell technologies, reflecting a move toward personalized medicine.


Subject(s)
Induced Pluripotent Stem Cells , Regenerative Medicine , Organoids , Tissue Engineering , Bibliometrics
2.
J Exp Med ; 220(11)2023 11 06.
Article in English | MEDLINE | ID: mdl-37725372

ABSTRACT

Accumulation of lipotoxic lipids, such as free cholesterol, induces hepatocyte death and subsequent inflammation and fibrosis in the pathogenesis of nonalcoholic steatohepatitis (NASH). However, the underlying mechanisms remain unclear. We have previously reported that hepatocyte death locally induces phenotypic changes in the macrophages surrounding the corpse and remnant lipids, thereby promoting liver fibrosis in a murine model of NASH. Here, we demonstrated that lysosomal cholesterol overload triggers lysosomal dysfunction and profibrotic activation of macrophages during the development of NASH. ß-cyclodextrin polyrotaxane (ßCD-PRX), a unique supramolecule, is designed to elicit free cholesterol from lysosomes. Treatment with ßCD-PRX ameliorated cholesterol accumulation and profibrotic activation of macrophages surrounding dead hepatocytes with cholesterol crystals, thereby suppressing liver fibrosis in a NASH model, without affecting the hepatic cholesterol levels. In vitro experiments revealed that cholesterol-induced lysosomal stress triggered profibrotic activation in macrophages predisposed to the steatotic microenvironment. This study provides evidence that dysregulated cholesterol metabolism in macrophages would be a novel mechanism of NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Mice , Disease Models, Animal , Liver Cirrhosis , Macrophages , Cholesterol , Lysosomes
3.
Nat Commun ; 14(1): 5534, 2023 09 25.
Article in English | MEDLINE | ID: mdl-37749092

ABSTRACT

Mesenchymal activation, characterized by dense stromal infiltration of immune and mesenchymal cells, fuels the aggressiveness of colorectal cancers (CRC), driving progression and metastasis. Targetable molecules in the tumor microenvironment (TME) need to be identified to improve the outcome in CRC patients with this aggressive phenotype. This study reports a positive link between high thrombospondin-1 (THBS1) expression and mesenchymal characteristics, immunosuppression, and unfavorable CRC prognosis. Bone marrow-derived monocyte-like cells recruited by CXCL12 are the primary source of THBS1, which contributes to the development of metastasis by inducing cytotoxic T-cell exhaustion and impairing vascularization. Furthermore, in orthotopically generated CRC models in male mice, THBS1 loss in the TME renders tumors partially sensitive to immune checkpoint inhibitors and anti-cancer drugs. Our study establishes THBS1 as a potential biomarker for identifying mesenchymal CRC and as a critical suppressor of antitumor immunity that contributes to the progression of this malignancy with a poor prognosis.


Subject(s)
Colorectal Neoplasms , Monocytes , Humans , Male , Animals , Mice , Immunosuppression Therapy , Aggression , Immune Checkpoint Inhibitors , Tumor Microenvironment
4.
Proc Natl Acad Sci U S A ; 120(33): e2304943120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37549290

ABSTRACT

Conventional dendritic cells (cDCs) are required for peripheral T cell homeostasis in lymphoid organs, but the molecular mechanism underlying this requirement has remained unclear. We here show that T cell-specific CD47-deficient (Cd47 ΔT) mice have a markedly reduced number of T cells in peripheral tissues. Direct interaction of CD47-deficient T cells with cDCs resulted in activation of the latter cells, which in turn induced necroptosis of the former cells. The deficiency and cell death of T cells in Cd47 ΔT mice required expression of its receptor signal regulatory protein α on cDCs. The development of CD4+ T helper cell-dependent contact hypersensitivity and inhibition of tumor growth by cytotoxic CD8+ T cells were both markedly impaired in Cd47 ΔT mice. CD47 on T cells thus likely prevents their necroptotic cell death initiated by cDCs and thereby promotes T cell survival and function.


Subject(s)
CD47 Antigen , CD8-Positive T-Lymphocytes , Animals , Mice , CD47 Antigen/genetics , CD47 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Survival , Dendritic Cells/metabolism , Necroptosis , Receptors, Immunologic/metabolism
6.
J Gastroenterol ; 58(5): 444-457, 2023 05.
Article in English | MEDLINE | ID: mdl-36739585

ABSTRACT

BACKGROUND: Amino acid transporters play an important role in supplying nutrition to cells and are associated with cell proliferation. L-type amino acid transporter 1 (LAT1) is highly expressed in many types of cancers and promotes tumor growth; however, how LAT1 affects tumor development is not fully understood. METHODS: To investigate the role of LAT1 in intestinal tumorigenesis, mice carrying LAT1 floxed alleles that also expressed Cre recombinase from the promoter of gene encoding Villin were crossed to an ApcMin/+ background (LAT1fl/fl; vil-cre; ApcMin/+), which were subject to analysis; organoids derived from those mice were also analyzed. RESULTS: This study showed that LAT1 was constitutively expressed in normal crypt base cells, and its conditional deletion in the intestinal epithelium resulted in fewer Paneth cells. LAT1 deletion reduced tumor size and number in the small intestine of ApcMin/+ mice. Organoids derived from LAT1-deleted ApcMin/+ intestinal crypts displayed fewer spherical organoids with reduced Wnt/ß-catenin target gene expression, suggesting a low tumor-initiation capacity. Wnt3 expression was decreased in the absence of LAT1 in the intestinal epithelium, suggesting that loss of Paneth cells due to LAT1 deficiency reduced the risk of tumor initiation by decreasing Wnt3 production. CONCLUSIONS: LAT1 affects intestinal tumor development in a cell-extrinsic manner through reduced Wnt3 expression in Paneth cells. Our findings may partly explain how nutrient availability can affect the risk of tumor development in the intestines.


Subject(s)
Adenomatous Polyposis Coli Protein , Amino Acid Transport System y+L , Intestinal Neoplasms , Paneth Cells , Animals , Mice , Cell Transformation, Neoplastic/genetics , Intestinal Mucosa/pathology , Intestinal Neoplasms/metabolism , Intestine, Small/pathology , Intestines , Paneth Cells/metabolism , Paneth Cells/pathology , Adenomatous Polyposis Coli Protein/metabolism , Amino Acid Transport System y+L/metabolism
7.
Cancer Sci ; 114(5): 1871-1881, 2023 May.
Article in English | MEDLINE | ID: mdl-36788737

ABSTRACT

Langerhans cell histiocytosis (LCH) is a rare neoplastic disorder characterized by inflammatory lesions arising from the anomalous accumulation of pathogenic CD1a+ CD207+ dendritic cells (DCs). SIRPα is a transmembrane protein highly expressed in myeloid cells such as DCs and macrophages. Here we show that SIRPα is a potential therapeutic target for LCH. We found that SIRPα is expressed in CD1a+ cells of human LCH lesions as well as in CD11c+ DCs in the spleen, liver, and lung of a mouse model of LCH (BRAFV600ECD11c mouse), in which an LCH-associated active form of human BRAF is expressed in a manner dependent on the mouse Cd11c promoter. BRAFV600ECD11c mice manifested markedly increased numbers of CD4+ T cells, regulatory T cells, and macrophages as well as of CD11c+ MHCII+ DCs in the spleen. Monotherapy with a mAb to SIRPα greatly reduced the percentage of CD11c+ MHCII+ DCs in peripheral blood, LCH-like lesion size in the liver, and the number of CD11c+ MHCII+ DCs in the spleen of the mutant mice. Moreover, this mAb promoted macrophage-mediated phagocytosis of CD11c+ DCs from BRAFV600ECD11c mice, whereas it had no effects on the viability or CCL19-dependent migration of such CD11c+ DCs or on their expression of the chemokine genes Ccl5, Ccl20, Cxcl11, and Cxcl12. Our results thus suggest that anti-SIRPα monotherapy is a promising approach to the treatment of LCH that is dependent in part on the promotion of the macrophage-mediated killing of LCH cells.


Subject(s)
Histiocytosis, Langerhans-Cell , Animals , Humans , Mice , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/metabolism , Spleen/metabolism
8.
Front Immunol ; 14: 1294814, 2023.
Article in English | MEDLINE | ID: mdl-38162643

ABSTRACT

Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment and are considered potential targets for cancer immunotherapy. To examine the antitumor effects of agents targeting human TAMs in vivo, we here established preclinical tumor xenograft models based on immunodeficient mice that express multiple human cytokines and have been reconstituted with a human immune system by transplantation of human CD34+ hematopoietic stem and progenitor cells (HIS-MITRG mice). HIS-MITRG mice supported the growth of both human cell line (Raji)- and patient-derived B cell lymphoma as well as the infiltration of human macrophages into their tumors. We examined the potential antitumor action of an antibody to human SIRPα (SE12C3) that inhibits the interaction of CD47 on tumor cells with SIRPα on human macrophages and thereby promotes Fcγ receptor-mediated phagocytosis of the former cells by the latter. Treatment with the combination of rituximab (antibody to human CD20) and SE12C3 inhibited Raji tumor growth in HIS-MITRG mice to a markedly greater extent than did rituximab monotherapy. This enhanced antitumor effect was dependent on human macrophages and attributable to enhanced rituximab-dependent phagocytosis of lymphoma cells by human macrophages. Treatment with rituximab and SE12C3 also induced reprogramming of human TAMs toward a proinflammatory phenotype. Furthermore, the combination treatment essentially prevented the growth of patient-derived diffuse large B cell lymphoma in HIS-MITRG mice. Our findings thus support the study of HIS-MITRG mice as a model for the preclinical evaluation in vivo of potential therapeutics, such as antibodies to human SIRPα, that target human TAMs.


Subject(s)
Antigens, Differentiation , Neoplasms , Humans , Mice , Animals , Rituximab/pharmacology , Rituximab/therapeutic use , Cell Line, Tumor , Antibodies , Immunotherapy , Disease Models, Animal , Neoplasms/therapy
9.
Cancers (Basel) ; 14(8)2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35454882

ABSTRACT

Conventional dendritic cells (cDCs) orchestrate immune responses to cancer and comprise two major subsets: type-1 cDCs (cDC1s) and type-2 cDCs (cDC2s). Compared with cDC1s, which are dedicated to the activation of CD8+ T cells, cDC2s are ontogenically and functionally heterogeneous, with their main function being the presentation of exogenous antigens to CD4+ T cells for the initiation of T helper cell differentiation. cDC1s play an important role in tumor-specific immune responses through cross-presentation of tumor-derived antigens for the priming of CD8+ T cells, whereas little is known of the role of cDC2s in tumor immunity. Recent studies have indicated that human cDC2s can be divided into at least two subsets and have implicated these cells in both anti- and pro-tumoral immune responses. Furthermore, the efficacy of cDC2-based vaccines as well as cDC2-targeted therapeutics has been demonstrated in both mouse models and human patients. Here we summarize current knowledge about the role of cDC2s in tumor immunity and address whether these cells are beneficial in the context of antitumor immune responses.

10.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34949714

ABSTRACT

The interaction of signal regulatory protein α (SIRPα) on macrophages with CD47 on cancer cells is thought to prevent antibody (Ab)-dependent cellular phagocytosis (ADCP) of the latter cells by the former. Blockade of the CD47-SIRPα interaction by Abs to CD47 or to SIRPα, in combination with tumor-targeting Abs such as rituximab, thus inhibits tumor formation by promoting macrophage-mediated ADCP of cancer cells. Here we show that monotherapy with a monoclonal Ab (mAb) to SIRPα that also recognizes SIRPß1 inhibited tumor formation by bladder and mammary cancer cells in mice, with this inhibitory effect being largely dependent on macrophages. The mAb to SIRPα promoted polarization of tumor-infiltrating macrophages toward an antitumorigenic phenotype, resulting in the killing and phagocytosis of cancer cells by the macrophages. Ablation of SIRPα in mice did not prevent the inhibitory effect of the anti-SIRPα mAb on tumor formation or its promotion of the cancer cell-killing activity of macrophages, however. Moreover, knockdown of SIRPß1 in macrophages attenuated the stimulatory effect of the anti-SIRPα mAb on the killing of cancer cells, whereas an mAb specific for SIRPß1 mimicked the effect of the anti-SIRPα mAb. Our results thus suggest that monotherapy with Abs to SIRPα/SIRPß1 induces antitumorigenic macrophages and thereby inhibits tumor growth and that SIRPß1 is a potential target for cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents/pharmacology , Immunotherapy/methods , Macrophages/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CD47 Antigen/metabolism , Cell Line, Tumor , Mice , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Rituximab , Treatment Outcome , Urinary Bladder
11.
Cell Stem Cell ; 29(2): 265-280.e6, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34856120

ABSTRACT

Adaptation to mechanical load, leading to enhanced force and power output, is a characteristic feature of skeletal muscle. Formation of new myonuclei required for efficient muscle hypertrophy relies on prior activation and proliferation of muscle stem cells (MuSCs). However, the mechanisms controlling MuSC expansion under conditions of increased load are not fully understood. Here we demonstrate that interstitial mesenchymal progenitors respond to mechanical load and stimulate MuSC proliferation in a surgical mouse model of increased muscle load. Mechanistically, transcriptional activation of Yes-associated protein 1 (Yap1)/transcriptional coactivator with PDZ-binding motif (Taz) in mesenchymal progenitors results in local production of thrombospondin-1 (Thbs1), which, in turn, drives MuSC proliferation through CD47 signaling. Under homeostatic conditions, however, CD47 signaling is insufficient to promote MuSC proliferation and instead depends on prior downregulation of the Calcitonin receptor. Our results suggest that relayed signaling between mesenchymal progenitors and MuSCs through a Yap1/Taz-Thbs1-CD47 pathway is critical to establish the supply of MuSCs during muscle hypertrophy.


Subject(s)
CD47 Antigen , Myoblasts , Animals , CD47 Antigen/metabolism , Hypertrophy/metabolism , Mice , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Stem Cells/metabolism
12.
PLoS One ; 16(8): e0256774, 2021.
Article in English | MEDLINE | ID: mdl-34437645

ABSTRACT

Cross talk between different signaling pathways is thought to be important for regulation of homeostasis of, as well as oncogenesis of, the intestinal epithelium. Expression of an active form of K-Ras specifically in intestinal epithelial cells (IECs) of mice (IEC-RasDA mice) resulted in the development of hyperplasia in the small intestine and colon of mice. IEC-RasDA mice also manifested the increased proliferation of IECs. In addition, the number of goblet cells markedly increased, while that of Paneth cells decreased in IEC-RasDA mice. Development of intestinal organoids was markedly enhanced for IEC-RasDA mice compared with control mice. Whereas, the expression of Wnt target genes was significantly reduced in the in intestinal crypts from IEC-RasDA mice compared with that apparent for the control. Our results thus suggest that K-Ras promotes the proliferation of IECs as well as generation of goblet cells. By contrast, Ras counter-regulates the Wnt signaling and thereby contribute to the proper regulation of intestinal epithelial cell homeostasis.


Subject(s)
Cell Proliferation/genetics , Homeostasis/genetics , Intestinal Mucosa/growth & development , Organoids/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Carcinogenesis/genetics , Colon/growth & development , Colon/pathology , Gene Expression Regulation, Neoplastic/genetics , Goblet Cells/metabolism , Humans , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Mice , Wnt Signaling Pathway/genetics
13.
Biochem Biophys Res Commun ; 569: 72-78, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34237430

ABSTRACT

The membrane protein SIRPα is a cold stress-responsive signaling molecule in neurons. Cold stress directly induces tyrosine phosphorylation of SIRPα in its cytoplasmic region, and phosphorylated SIRPα is involved in regulating experience-dependent behavioral changes in mice. Here, we examined the mechanism of cold stress-induced SIRPα phosphorylation in vitro and in vivo. The levels of activated Src family protein tyrosine kinases (SFKs), which phosphorylate SIRPα, were not increased by lowering the temperature in cultured neurons. Although the SFK inhibitor dasatinib markedly reduced SIRPα phosphorylation, low temperature induced an increase in SIRPα phosphorylation even in the presence of dasatinib, suggesting that SFK activation is not required for low temperature-induced SIRPα phosphorylation. However, in the presence of pervanadate, a potent inhibitor of protein tyrosine phosphatases (PTPases), SIRPα phosphorylation was significantly reduced by lowering the temperature, suggesting that either the inactivation of PTPase(s) that dephosphorylate SIRPα or increased protection of phosphorylated SIRPα from the PTPase activity is important for low temperature-induced SIRPα phosphorylation. Inactivation of PTPase Shp2 by the allosteric Shp2 inhibitor SHP099, but not by the competitive inhibitor NSC-87877, reduced SIRPα phosphorylation in cultured neurons. Shp2 knockout also reduced SIRPα phosphorylation in the mouse brain. Our data suggest that Shp2, but not SFKs, positively regulates cold stress-induced SIRPα phosphorylation in a PTPase activity-independent manner.


Subject(s)
Cold Temperature , Neurons/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptors, Immunologic/metabolism , Tyrosine/metabolism , Animals , Cells, Cultured , Cold-Shock Response , Dasatinib/pharmacology , Immunoblotting , Mice, Knockout , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Phosphorylation/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Pyrimidines/pharmacology
14.
Cancer Sci ; 112(1): 16-21, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33073467

ABSTRACT

The turnover of intestinal epithelial cells (IECs) is relatively rapid (3-5 days in mouse and human), and this short existence and other aspects of the homeostasis of IECs are tightly regulated by various signaling pathways including Wnt-ß-catenin signaling. Dysregulation of IEC homeostasis likely contributes to the development of intestinal inflammation and intestinal cancer. The roles of receptor protein tyrosine kinases and their downstream signaling molecules such as Src family kinases, Ras, and mTOR in homeostatic regulation of IEC turnover have recently been evaluated. These signaling pathways have been found to promote not only the proliferation of IECs but also the differentiation of progenitor cells into secretory cell types such as goblet cells. Of note, signaling by Src family kinases, Ras, and mTOR has been shown to oppose the Wnt-ß-catenin signaling pathway and thereby to limit the number of Lgr5+ intestinal stem cells or of Paneth cells. Such cross-talk of signaling pathways is important not only for proper regulation of IEC homeostasis but for the development of intestinal tumors and potentially for anticancer therapy.


Subject(s)
Carcinogenesis/metabolism , Epithelial Cells/metabolism , Homeostasis/physiology , Intestines/pathology , TOR Serine-Threonine Kinases/metabolism , ras Proteins/metabolism , src-Family Kinases/metabolism , Animals , Carcinogenesis/pathology , Epithelial Cells/pathology , Humans
15.
Front Immunol ; 11: 570963, 2020.
Article in English | MEDLINE | ID: mdl-33162986

ABSTRACT

The inhibitory immunoreceptor SIRPα is expressed on myeloid and neuronal cells and interacts with the broadly expressed CD47. CD47-SIRPα interactions form an innate immune checkpoint and its targeting has shown promising results in cancer patients. Here, we report expression of SIRPα on B1 lymphocytes, a subpopulation of murine B cells responsible for the production of natural antibodies. Mice defective in SIRPα signaling (SIRPαΔCYT mice) displayed an enhanced CD11b/CD18 integrin-dependent B1 cell migration from the peritoneal cavity to the spleen, local B1 cell accumulation, and enhanced circulating natural antibody levels, which was further amplified upon immunization with T-independent type 2 antigen. As natural antibodies are atheroprotective, we investigated the involvement of SIRPα signaling in atherosclerosis development. Bone marrow (SIRPαΔCYT>LDLR-/-) chimaeric mice developed reduced atherosclerosis accompanied by increased natural antibody production. Collectively, our data identify SIRPα as a unique B1 cell inhibitory receptor acting to control B1 cell migration, and imply SIRPα as a potential therapeutic target in atherosclerosis.


Subject(s)
Atherosclerosis/immunology , B-Lymphocytes/immunology , CD47 Antigen/metabolism , Lymphoid Tissue/immunology , Receptors, Immunologic/metabolism , Animals , Antibody Formation , Autoantibodies/metabolism , Cell Movement , Cells, Cultured , Cytokines/metabolism , Immunomodulation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics , Receptors, LDL/genetics , Th1 Cells/immunology , Transplantation Chimera
16.
Expert Opin Ther Targets ; 24(10): 945-951, 2020 10.
Article in English | MEDLINE | ID: mdl-32799682

ABSTRACT

The CD47-Signal regulatory protein α (SIRPα) singling axis acts as a crucial regulator that limits the phagocytic activity of professional phagocytes such as macrophages. Recent studies have demonstrated that the interaction between CD47 on tumor cells and SIRPα on macrophages is implicated in the ability of tumors to evade immunosurveillance. Targeting the CD47-SIRPα interaction is therefore considered to be a promising approach for cancer therapy. Herein, we review some of studies displaying the potential clinical application of antibodies and other modalities that target the CD47-SIRPα interaction. Current limitations of the CD47-SIRPα-targeted immunotherapeutic approaches are also discussed as well as other avenues for future study to improve the current strategies in targeting the CD47-SIRPα signaling axis for cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Molecular Targeted Therapy , Neoplasms/therapy , Animals , Antibodies/immunology , Antigens, Differentiation/immunology , CD47 Antigen/immunology , Humans , Macrophages/metabolism , Neoplasms/immunology , Receptors, Immunologic/immunology
17.
Sci Rep ; 10(1): 13810, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796887

ABSTRACT

Cell signaling important for homeostatic regulation of colonic epithelial cells (CECs) remains poorly understood. Mammalian target of rapamycin complex 1 (mTORC1), a protein complex that contains the serine-threonine kinase mTOR, mediates signaling that underlies the control of cellular functions such as proliferation and autophagy by various external stimuli. We here show that ablation of tuberous sclerosis complex 2 (Tsc2), a negative regulator of mTORC1, specifically in intestinal epithelial cells of mice resulted in increased activity of mTORC1 of, as well as increased proliferative activity of, CECs. Such Tsc2 ablation also reduced the population of Lgr5-positive colonic stem cells and the expression of Wnt target genes in CECs. The stimulatory phosphorylation of the kinase Akt and inhibitory phosphorylation of glycogen synthase kinase 3ß were both markedly decreased in the colon of the Tsc2 conditional knockout (CKO) mice. Development of colonic organoids with cryptlike structures was enhanced for Tsc2 CKO mice compared with control mice. Finally, Tsc2 CKO mice manifested increased susceptibility to dextran sulfate sodium-induced colitis. Our results thus suggest that mTORC1 activity promotes the proliferation of, as well as the expression of Wnt target genes in, CECs and thereby contributes to colonic organogenesis and homeostasis.


Subject(s)
Cell Proliferation/genetics , Colitis/genetics , Colon/cytology , Epithelial Cells/physiology , Homeostasis/genetics , Mechanistic Target of Rapamycin Complex 1/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Animals , Autophagy/genetics , Cell Proliferation/physiology , Cells, Cultured , Genetic Predisposition to Disease , Glycogen Synthase Kinase 3 beta/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Knockout , Phosphorylation , Tuberous Sclerosis Complex 2 Protein/physiology
18.
Cell Chem Biol ; 27(9): 1181-1191.e7, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32640189

ABSTRACT

Medium-sized macrocyclic peptides are an alternative to small compounds and large biomolecules as a class of pharmaceutics. The CD47-SIRPα signaling axis functions as an innate immune checkpoint that inhibits phagocytosis in phagocytes and has been implicated as a promising target for cancer immunotherapy. The potential of macrocyclic peptides that target this signaling axis as immunotherapeutic agents has remained unknown, however. Here we have developed a macrocyclic peptide consisting of 15 amino acids that binds to the ectodomain of mouse SIRPα and efficiently blocks its interaction with CD47 in an allosteric manner. The peptide markedly promoted the phagocytosis of antibody-opsonized tumor cells by macrophages in vitro as well as enhanced the inhibitory effect of anti-CD20 or anti-gp75 antibodies on tumor formation or metastasis in vivo. Our results suggest that allosteric inhibition of the CD47-SIRPα interaction by macrocyclic peptides is a potential approach to cancer immunotherapy.


Subject(s)
CD47 Antigen/metabolism , Peptides, Cyclic/metabolism , Receptors, Immunologic/metabolism , Allosteric Regulation , Animals , Antigens, CD20/immunology , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , CD47 Antigen/chemistry , Cell Line, Tumor , Female , Humans , Immunotherapy , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Peptides, Cyclic/chemistry , Peptides, Cyclic/therapeutic use , Phagocytosis , Protein Binding , Receptors, Immunologic/chemistry , Rituximab/immunology , Rituximab/therapeutic use , Survival Rate
19.
PLoS Pathog ; 16(7): e1008609, 2020 07.
Article in English | MEDLINE | ID: mdl-32702057

ABSTRACT

Primary infection of human herpesvirus 6B (HHV-6B) occurs in infants after the decline of maternal immunity and causes exanthema subitum accompanied by a high fever, and it occasionally develops into encephalitis resulting in neurological sequelae. There is no effective prophylaxis for HHV-6B, and its development is urgently needed. The glycoprotein complex gH/gL/gQ1/gQ2 (called 'tetramer of HHV-6B') on the virion surface is a viral ligand for its cellular receptor human CD134, and their interaction is thus essential for virus entry into the cells. Herein we examined the potency of the tetramer as a vaccine candidate against HHV-6B. We designed a soluble form of the tetramer by replacing the transmembrane domain of gH with a cleavable tag, and the tetramer was expressed by a mammalian cell expression system. The expressed recombinant tetramer is capable of binding to hCD134. The tetramer was purified to homogeneity and then administered to mice with aluminum hydrogel adjuvant and/or CpG oligodeoxynucleotide adjuvant. After several immunizations, humoral and cellular immunity for HHV-6B was induced in the mice. These results suggest that the tetramer together with an adjuvant could be a promising candidate HHV-6B vaccine.


Subject(s)
Exanthema Subitum/immunology , Herpesvirus Vaccines/immunology , Viral Envelope Proteins/immunology , Adjuvants, Immunologic/pharmacology , Animals , Exanthema Subitum/virology , Herpesvirus 6, Human , Humans , Mice , Mice, Inbred BALB C
20.
Eur J Immunol ; 50(10): 1560-1570, 2020 10.
Article in English | MEDLINE | ID: mdl-32438469

ABSTRACT

Signal regulatory protein α (SIRPα) is expressed predominantly on type 2 conventional dendritic cells (cDC2s) and macrophages. We previously showed that mice systemically lacking SIRPα were resistant to experimental autoimmune encephalomyelitis (EAE). Here, we showed that deletion of SIRPα in CD11c+ cells of mice (SirpaΔDC mice) also markedly ameliorated the development of EAE. The frequency of cDCs and migratory DCs (mDCs), as well as that of Th17 cells, were significantly reduced in draining lymph nodes of SirpaΔDC mice at the onset of EAE. In addition, we found the marked reduction in the number of Th17 cells and DCs in the CNS of SirpaΔDC mice at the peak of EAE. Whereas inducible systemic ablation of SIRPα before the induction of EAE prevented disease development, that after EAE onset did not ameliorate the clinical signs of disease. We also found that EAE development was partially attenuated in mice with CD11c+ cell-specific ablation of CD47, a ligand of SIRPα. Collectively, our results suggest that SIRPα expressed on CD11c+ cells, such as cDC2s and mDCs, is indispensable for the development of EAE, being required for the priming of self-reactive Th17 cells in the periphery as well as for the inflammation in the CNS.


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Multiple Sclerosis/immunology , Receptors, Immunologic/metabolism , Th17 Cells/immunology , Animals , CD11c Antigen/metabolism , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Immunologic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...