scholarly journals Anti-sortilin1 Antibody Up-Regulates Progranulin via Sortilin1 Down-Regulation

2020 ◽  
Vol 14 ◽  
Author(s):  
Shuuichi Miyakawa ◽  
Hiroyuki Sakuma ◽  
Dnyaneshwar Warude ◽  
Satomi Asanuma ◽  
Naoto Arimura ◽  
...  

Progranulin (PGRN) haploinsufficiency associated with loss-of-function mutations in the granulin gene causes frontotemporal dementia (FTD). This suggests that increasing PGRN levels could have promising therapeutic implications for patients carrying GRN mutations. In this study, we explored the therapeutic potential of sortilin1 (SORT1), a clearance receptor of PGRN, by generating and characterizing monoclonal antibodies against SORT1. Anti-SORT1 monoclonal antibodies were generated by immunizing Sort1 knockout mice with SORT1 protein. The antibodies were classified into 7 epitope bins based on their competitive binding to the SORT1 protein and further defined by epitope bin-dependent characteristics, including SORT1-PGRN blocking, SORT1 down-regulation, and binding to human and mouse SORT1. We identified a positive correlation between PGRN up-regulation and SORT1 down-regulation. Furthermore, we also characterized K1-67 antibody via SORT1 down-regulation and binding to mouse SORT1 in vivo and confirmed that K1-67 significantly up-regulated PGRN levels in plasma and brain interstitial fluid of mice. These data indicate that SORT1 down-regulation is a key mechanism in increasing PGRN levels via anti-SORT1 antibodies and suggest that SORT1 is a potential target to correct PGRN reduction, such as that in patients with FTD caused by GRN mutation.

Blood ◽  
2007 ◽  
Vol 110 (2) ◽  
pp. 529-535 ◽  
Author(s):  
Tamer Rabie ◽  
David Varga-Szabo ◽  
Markus Bender ◽  
Rastislav Pozgaj ◽  
Francois Lanza ◽  
...  

Abstract Coronary artery thrombosis is often initiated by platelet activation on collagen-rich subendothelial layers in the disrupted atherosclerotic plaque. The activating platelet collagen receptor glycoprotein VI (GPVI) noncovalently associates with the Fc receptor γ-chain (FcRγ), which signals through its immunoreceptor-tyrosine–based activation motif (ITAM) via the adaptor LAT leading to the activation of phospholipase Cγ2 (PLCγ2). GPVI is a promising antithrombotic target as anti-GPVI antibodies induce the irreversible loss of the receptor from circulating platelets by yet undefined mechanisms in humans and mice and long-term antithrombotic protection in the latter. However, the treatment is associated with transient but severe thrombocytopenia and reduced platelet reactivity to thrombin questioning its clinical usefulness. Here we show that GPVI down-regulation occurs through 2 distinct pathways, namely ectodomain shedding or internalization/intracellular clearing, and that both processes are abrogated in mice carrying a point mutation in the FcRγ-associated ITAM. In mice lacking LAT or PLCγ2, GPVI shedding is abolished, but the receptor is irreversibly down-regulated through internalization/intracellular clearing. This route of GPVI loss is not associated with thrombocytopenia or altered thrombin responses. These results reveal the existence of 2 distinct signaling pathways downstream of the FcRγ-ITAM and show that it is possible to uncouple GPVI down-regulation from undesired side effects with obvious therapeutic implications.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 938-938
Author(s):  
Shujun Liu ◽  
Lai Chu Wu ◽  
Jiuxia Pang ◽  
Ramasamy Santhanam ◽  
Sebastian Schwind ◽  
...  

Abstract Abstract 938 KIT is a receptor tyrosine kinase (RTK) and its aberrant activities resulting from protein overexpression and/or activating mutations are associated with a number of malignancies including core binding factor (CBF) AML [e.g., patients with t(8;21) or inv(16) or molecular equivalent RUNX1/RUNX1T1 or CBFB/MYH11, respectively]. RTK inhibitors (e.g. PKC412) have been shown to suppress aberrant KIT activity and delay tumor growth, but they are active only on distinct types of KIT mutations (KITmut). Furthermore, resistance to these inhibitors, as a result of secondary mutations or KIT overexpression, is emerging. Thus, we hypothesize that direct inhibition of KIT gene transcription may be a valuable therapeutic approach to override aberrant KIT expression and activity. Here, we described the regulatory and functional role of Sp1/NFkB-miR29b feedback loop in KIT-driven leukemia that can be targeted pharmacologically. Applying chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays (EMSA) to RUNX1/RUNX1T1-positive Kasumi-1 cells, we demonstrated that while, the Sp1/NFkB complex was enriched on KIT promoter and acted as gene transactivator thereby leading to KIT overexpression, Sp1/NFkB recruited HDAC1 and HDAC3 to miR29b regulatory region thereby epigenetically repressing miR29b. This microRNA, when expressed, targeted Sp1 and eventually decreased Sp1/NFkB-mediated gene transactivation, including that of KIT. In agreement with these, we showed that when Sp1, NFkB and HDAC1 were transiently overexpressed in Kasumi-1 cells, increased KIT expression and decreased miR29b transcription occurred. In contrast, siRNA knockdown of Sp1, NFkB and HDAC1 augmented miR29b level and decreased KIT transcription. Moreover, ectopic miR29b expression impaired Sp1/NFkB repressor complex on the promoter of endogenous miR29b, thereby resulting in re-expression of the endogenous microRNA and further inhibition of Sp1/NFkB-dependent KIT transcription. Importantly, the activity of Sp1/NFkB/HDACs complex can also be pharmacologically modulated leading to restored miR29b transcription and abrogated KIT expression. We showed that pharmacologic interference with Sp1/NFkB/HDACs using their respective inhibitors, such as bortezomib (0, 6, 20, 60 and 100nM for Sp1 and NFkB), mithramycin A (150 and 300ng/ml for Sp1), bay 11-7082 (3μM for NFkB) and OSU-HDAC42 (1μM for HDAC), upregulated miR29b at early time point (6 hours) and decreased Sp1 and in turn KIT expression in KIT overexpression cell lines (e.g., Kasumi-1) and AML patient blasts. EMSA and ChIP assay demonstrated that bortezomib or HDAC42-mediated KIT repression and miR29b upregulation occurred through the dissociation of Sp1/NFkB complex from the corresponding promoter. To further investigate the therapeutic potential of targeting KIT over-expression in leukemia, we stably expressed KIT wild type (KITwt) or KITmut (D816V) in FDC-P1 cell line (murine non-tumorigenic cells derived from myeloid precursors), and we evidenced that both KITwt and KITmut promoted cell proliferation that was overcome by bortezomib in clonogenic assay. In in vivo study, when NOD/SCID mice were engrafted with FDC-P1/KITmut cells (5×106/mouse), they developed significant splenomegaly and marrow blast infiltration through KIT overexpression. When leukemia-carrying mice were treated with bortezomib (1mg/kg) for 48 hours, we observed an obvious increase of endogenous miR29b expression and a significant reduction of KIT expression. Leukemic mice that received 1mg/kg of bortezomib twice/week for 3 weeks starting on day 21 after engraftment (n=5 mice/group) showed no evidence of splenomegaly and had a significantly longer median survival [59 days (twice/week) vs 28 days (vehicle-treated), p=0.0036], compared to vehicle-treated mice that instead showed massive splenomegaly. Cytospin of marrow and histopathology of spleen and liver showed that vehicle-treated mice displayed extensive blast infiltration that was instead absent in bortezomib-treated mice. Altogether, our study revealed a previously unrecognized protein-microRNA regulatory network whose imbalance contributes to KIT-driven leukemia. As the aberrant activity of this network is pharmacologically targetable, this discovery may be quickly translated into the clinic as a novel therapeutic approach for KIT-driven AML and other malignancies. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 2009 ◽  
pp. 1-10 ◽  
Author(s):  
Helenia Ansuini ◽  
Annalisa Meola ◽  
Zeynep Gunes ◽  
Valentina Paradisi ◽  
Monica Pezzanera ◽  
...  

The EphA2 receptor tyrosine kinase is overexpressed in a variety of human epithelial cancers and is a determinant of malignant cellular behavior in pancreatic adenocarcinoma cells. Moreover, it is expressed in tumor endothelium and its activation promotes angiogenesis. To better clarify the therapeutic potential of monoclonal antibodies (mAbs) directed to the EphA2 receptor, we generated a large number of mAbs by differential screening of phage-Ab libraries by oligonucleotide microarray technology and implemented a strategy for the rapid identification of antibodies with the desired properties. We selected two high-affinity and highly specific EphA2 monoclonal antibodies with different in vitro properties on the human pancreatic tumor cell line MiaPaCa2. One is a potent EphA2-agonistic antibody, IgG25, that promotes receptor endocytosis and subsequent degradation, and the second is a ligand antagonist, IgG28, that blocks the binding to ephrin A1 and is cross-reactive with the mouse EphA2 receptor. We measured the effect of antibody treatment on the growth of MiaPaCa2 cells orthotopically transplanted in nude mice. Both IgG25 and IgG28 had strong antitumor and antimetastatic efficacy. In vivo treatment with IgG25 determined the reduction of the EphA2 protein levels in the tumor and the phosphorylation of FAK on Tyr576 while administration of IgG28 caused a decrease in tumor vascularization as measured by immunohistochemical analysis of CD31 in tumor sections. These data show that in a pancreatic cancer model comparable therapeutic efficacy is obtained either by promoting receptor degradation or by blocking receptor activation.


2021 ◽  
Vol 13 (584) ◽  
pp. eaay9056 ◽  
Author(s):  
Ana M. Moreno ◽  
Fernando Alemán ◽  
Glaucilene F. Catroli ◽  
Matthew Hunt ◽  
Michael Hu ◽  
...  

Current treatments for chronic pain rely largely on opioids despite their substantial side effects and risk of addiction. Genetic studies have identified in humans key targets pivotal to nociceptive processing. In particular, a hereditary loss-of-function mutation in NaV1.7, a sodium channel protein associated with signaling in nociceptive sensory afferents, leads to insensitivity to pain without other neurodevelopmental alterations. However, the high sequence and structural similarity between NaV subtypes has frustrated efforts to develop selective inhibitors. Here, we investigated targeted epigenetic repression of NaV1.7 in primary afferents via epigenome engineering approaches based on clustered regularly interspaced short palindromic repeats (CRISPR)–dCas9 and zinc finger proteins at the spinal level as a potential treatment for chronic pain. Toward this end, we first optimized the efficiency of NaV1.7 repression in vitro in Neuro2A cells and then, by the lumbar intrathecal route, delivered both epigenome engineering platforms via adeno-associated viruses (AAVs) to assess their effects in three mouse models of pain: carrageenan-induced inflammatory pain, paclitaxel-induced neuropathic pain, and BzATP-induced pain. Our results show effective repression of NaV1.7 in lumbar dorsal root ganglia, reduced thermal hyperalgesia in the inflammatory state, decreased tactile allodynia in the neuropathic state, and no changes in normal motor function in mice. We anticipate that this long-lasting analgesia via targeted in vivo epigenetic repression of NaV1.7 methodology we dub pain LATER, might have therapeutic potential in management of persistent pain states.


2019 ◽  
Vol 3 (15) ◽  
pp. 2355-2367 ◽  
Author(s):  
Yueh-Chwen Hsu ◽  
Tsung-Chih Chen ◽  
Chien-Chin Lin ◽  
Chang-Tsu Yuan ◽  
Chia-Lang Hsu ◽  
...  

Abstract Plant homeodomain finger gene 6 (PHF6) encodes a 365-amino-acid protein containing 2 plant homology domain fingers. Germline mutations of human PHF6 cause Börjeson-Forssman-Lehmann syndrome, a congenital neurodevelopmental disorder. Loss-of-function mutations of PHF6 are detected in patients with acute leukemia, mainly of T-cell lineage and in a small proportion of myeloid lineage. The functions of PHF6 in physiological hematopoiesis and leukemogenesis remain incompletely defined. To address this question, we generated a conditional Phf6 knockout mouse model and investigated the impact of Phf6 loss on the hematopoietic system. We found that Phf6 knockout mice at 8 weeks of age had reduced numbers of CD4+ and CD8+ T cells in the peripheral blood compared with the wild-type littermates. There were decreased granulocyte-monocytic progenitors but increased Lin–c-Kit+Sca-1+ cells in the marrow of young Phf6 knockout mice. Functional studies, including competitive repopulation unit and serial transplantation assays, revealed an enhanced reconstitution and self-renewal capacity in Phf6 knockout hematopoietic stem cells (HSCs). Aged Phf6 knockout mice had myelodysplasia-like presentations, including decreased platelet counts, megakaryocyte dysplasia, and enlarged spleen related to extramedullary hematopoiesis. Moreover, we found that Phf6 loss lowered the threshold of NOTCH1-induced leukemic transformation at least partially through increased leukemia-initiating cells. Transcriptome analysis on the restrictive rare HSC subpopulations revealed upregulated cell cycling and oncogenic functions, with alteration of key gene expression in those pathways. In summary, our studies show the in vivo crucial roles of Phf6 in physiological and malignant hematopoiesis.


2020 ◽  
Vol 5 (54) ◽  
pp. eaax1686
Author(s):  
Zaruhi Hovhannisyan ◽  
Nengyin Liu ◽  
Sara Khalil-Aguero ◽  
Casandra Panea ◽  
Jeffrey VanValkenburgh ◽  
...  

Deficiency in interleukin-36R (IL-36R) antagonist caused by loss-of-function mutations in IL-36RN leads to DITRA (deficiency of IL-36 receptor antagonist), a rare inflammatory human disease that belongs to a subgroup of generalized pustular psoriasis (GPP). We report a functional genetic mouse model of DITRA with enhanced IL-36R signaling analogous to that observed in patients with DITRA, which provides new insight into our understanding of the IL-36 family of molecules in regulating barrier integrity across multiple tissues. Humanized DITRA-like mice displayed increased skin inflammation in a preclinical model of psoriasis, and in vivo blockade of IL-36R pathway using anti-human IL-36R antibody ameliorated imiquimod-induced skin pathology as both prophylactic and therapeutic treatments. Deeper characterization of the humanized DITRA-like mice revealed that deregulated IL-36R signaling promoted tissue pathology during intestinal injury and led to impairment in mucosal restoration in the repair phase of chronic dextran sulfate sodium (DSS)–induced colitis. Blockade of IL-36R pathway significantly ameliorated DSS-induced intestinal inflammation and rescued the inability of DITRA-like mice to recover from mucosal damage in vivo. Our results indicate a central role for IL-36 in regulating proinflammatory responses in the skin and epithelial barrier function in the intestine, suggesting a new therapeutic potential for targeting the IL-36R axis in psoriasis and at the later stages of intestinal pathology in inflammatory bowel disease.


2021 ◽  
Author(s):  
Moataz Dowaidar

Tissue hypoxia has been found as a master regulator of alternative splicing, which can have significant clinical implications. Hypoxia-elicited AS is more common in the setting of various cancer hallmarks than other illnesses, owing to the fact that hypoxia and AS are intensively explored in cancer. However, an increasing number of hypoxia-induced AS episodes have been linked to a variety of clinical conditions, including neurological and cardiovascular disorders. Hypoxia-induced AS, of course, has its own set of markers with prognostic and therapeutic implications. Targeted regulation of hypoxia signaling with the objective of modulating hypoxia-driven AS is of great interest in some cancers. In order to design acceptable therapeutic paradigms, future research will be necessary to unravel the proper molecular pathways. Although some of the discovered molecular targets appear to have therapeutic potential, more in-vivo research is required.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 833-833
Author(s):  
Sophia Adamia ◽  
Mariateresa Fulciniti ◽  
Herve Avet-Loiseau ◽  
Samir B Amin ◽  
Parantu Shah ◽  
...  

Abstract Abstract 833 A growing body of evidence suggests that the genome of a many organisms, particularly mammals is controlled not only by transcription factors but also by post-transcriptional programs that are modulated by the family of small RNA molecules including microRNAs (miRs). miRs can block mRNA translation and affect mRNA stability. We have evaluated profiles of 384 human miRs in CD138+ cells from 79 patients with multiple myeloma (MM), 11 MM cell lines and 9 healthy donors (HD) using qRT-PCR based microRNA array. This analysis has identified a MM specific miRNA signature that significantly correlates with OS (p=0.05) and EFS (p=0.017) of patients. Based on this signature one group of patients clustered with HD suggesting indolent disease while other with cell lines indicating aggressive disease. We identified significant modulation of expression of 61 microRNAs in MM cells compared to normal plasma cells. Specific miRs with established oncogenic and tumor suppressor functions such as miR-155, miR-585 and Let7-f were significantly dysregulated in MM (p<0.001). Modulation of miRs-155, -585 and Let7 were observed most frequently in the group of patients with poor OS and EFS suggesting their crucial role in MM. However biological role of these miRs have not yet been defined. To further evaluate biological function of these most recurrent miRs in MM, we evaluated role of miR-155, let-7f and mir-585 in MM cell lines by gain- and loss- of function experiments. We used locked nucleic acid (LNA) anti-miR probes for loss of function and pre-miR-155 for gain of function studies using them alone or in combination. Although manipulation of all 3 miRs induced 20-25% change in MM cell proliferation and/or induction of apoptosis, combination of anti-miR-let7f with pre-miR-155, and anti-miR-585 in combination with miR-155 had dramatic effects on MM cell proliferation and over 60% cells undergoing apoptosis. To evaluate the targets of these miRs, we have determined effects of these anti-miRs and pre-miR on global gene and miR expression profile in MM alone and in combinations. This analysis identified modulation of cluster of miRs as well as genes critical for cell growth and survival. Next, we have tested efficacy of these miRs in vivo in murine Xenograft model to evaluate their therapeutic potential. Tumor-bearing mice were treated intraperitoneal for four consecutively days with the LNA anti-miR-585 and Let-7 and pre-miR-155 probes and respective controls alone and in combination. We observed that the single LNA anti-miR-585 and let 7 and pre miR-155 treatment reduced tumor size by 36%, 31% and 155% in animal 7 days after treatment. However, significant tumor size reductions were achieved when animals were treated with combinations; anti-miR-Let 7f plus pre-miR-155 (58 %); LNA anti-miR-Let 7f plus LNA anti-miR-585 (56 %); LNA-anti-miR-585 plus pre-miR-155 (74 %).We did not observe any significant systemic toxicity in the animals. In conclusion our results suggest significant biological role for miR-585, let 7f and miR-155 in myeloma, both in vitro and in vivo; it highlights for the first time a concerted activity of combination of miRs and holds a great promise for developing novel therapeutic approach for myeloma. Disclosures: No relevant conflicts of interest to declare.


Endocrinology ◽  
2009 ◽  
Vol 150 (9) ◽  
pp. 4163-4169 ◽  
Author(s):  
Shengjun Qiao ◽  
Sam Okret ◽  
Mikael Jondal

Abstract Thymocytes from adult mice synthesize glucocorticoids (GCs), and some data indicate a role for this hormone production in thymic homeostasis. Here we present further support for this view by showing that the dramatic increase in thymocyte number seen after adrenalectomy (ADX) does not correlate with the decrease in systemic GCs but rather with an ACTH-mediated down-regulation of GC synthesis in thymocytes. High ACTH concentrations caused by ADX in wild-type mice down-regulated CYP11B1 mRNA expression, encoding the last enzyme required for corticosterone synthesis and as a consequence reduced GC synthesis in thymocytes. This was not seen in IL-1β/IL-18 double-knockout mice unable to respond to ADX with high ACTH levels. However, if ADX IL-1β/IL-18 double-knockout mice were treated with ACTH, this led to a down-regulation of CYP11B1 and GC synthesis in thymocytes. In addition, in vivo treatment of mice with the CYP11B1 antagonist metyrapone, without affecting the systemic corticosterone level, increased thymocyte numbers and in vitro treatment of isolated thymocytes prevented thymocyte loss. Furthermore, in vitro experiments showed that both ACTH and its receptor-induced second-messenger molecule cAMP down-regulated mRNA expression of critical enzymes in GC steroidogenesis and GC synthesis in thymocytes. We conclude that thymocyte-produced GCs are important for the homeostasis of adult mouse thymocytes and that high ACTH level, in contrast to stimulating GC synthesis in the adrenal glands, has the opposite effect in thymocytes.


2008 ◽  
Vol 82 (14) ◽  
pp. 7009-7021 ◽  
Author(s):  
Ana P. Goncalvez ◽  
Cheng-Hsin Chien ◽  
Kamolchanok Tubthong ◽  
Inna Gorshkova ◽  
Carrie Roll ◽  
...  

ABSTRACT Japanese encephalitis virus (JEV)-specific Fab antibodies were recovered by repertoire cloning from chimpanzees initially immunized with inactivated JE-VAX and then boosted with attenuated JEV SA14-14-2. From a panel of 11 Fabs recovered by different panning strategies, three highly potent neutralizing antibodies, termed Fabs A3, B2, and E3, which recognized spatially separated regions on the virion, were identified. These antibodies reacted with epitopes in different domains: the major determinant for Fab A3 was Lys179 (domain I), that for Fab B2 was Ile126 (domain II), and that for Fab E3 was Gly302 (domain III) in the envelope protein, suggesting that these antibodies neutralize the virus by different mechanisms. Potent neutralizing antibodies reacted with a low number of binding sites available on the virion. These three Fabs and derived humanized monoclonal antibodies (MAbs) exhibited high neutralizing activities against a broad spectrum of JEV genotype strains. Demonstration of antibody-mediated protection of JEV infection in vivo is provided using the mouse encephalitis model. MAb B2 was most potent, with a 50% protective dose (ED50) of 0.84 μg, followed by MAb A3 (ED50 of 5.8 μg) and then MAb E3 (ED50 of 24.7 μg) for a 4-week-old mouse. Administration of 200 μg/mouse of MAb B2 1 day after otherwise lethal JEV infection protected 50% of mice and significantly prolonged the average survival time compared to that of mice in the unprotected group, suggesting a therapeutic potential for use of MAb B2 in humans.


Sign in / Sign up

Export Citation Format

Share Document