scholarly journals AHR:IKAROS Interaction Promotes Platelet Biogenesis in Response to SR1

Reports ◽  
2021 ◽  
Vol 4 (1) ◽  
pp. 7
Author(s):  
Lea Mallo ◽  
Valentin Do Sacramento ◽  
Christian Gachet ◽  
Susan Chan ◽  
Philippe Kastner ◽  
...  

In vitro, the differentiation of megakaryocytes (MKs) is improved by aryl-hydrocarbon receptor (AHR) antagonists such as StemRegenin 1 (SR1), an effect physiologically recapitulated by the presence of stromal mesenchymal cells (MSC). This inhibition promotes the amplification of a CD34+CD41low population able to mature as MKs with a high capacity for platelet production. In this short report, we showed that the emergence of the thrombocytogenic precursors and the enhancement of platelet production triggered by SR1 involved IKAROS. The downregulation/inhibition of IKAROS (shRNA or lenalidomide) significantly reduced the emergence of SR1-induced thrombocytogenic population, suggesting a crosstalk between AHR and IKAROS. Interestingly, using a proximity ligation assay, we could demonstrate a physical interaction between AHR and IKAROS. This interaction was also observed in the megakaryocytic cells differentiated in the presence of MSCs. In conclusion, our study revealed a previously unknown AHR/ IKAROS -dependent pathway which prompted the expansion of the thrombocytogenic precursors. This AHR- IKAROS dependent checkpoint controlling MK maturation opens new perspectives to platelet production engineering.

2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Gunsmaa Nyamsuren ◽  
Gregor Christof Rapp ◽  
Björn Tampe ◽  
Michael Zeisberg

Abstract Background and Aims Aryl hydrocarbon receptor nuclear translocator (ARNT) mediates anti-fibrotic activity in kidney and liver through induction of ALK3-receptor expression and subsequently increased Smad1/5/8 signaling. While expression of ARNT can be pharmacologically induced by sub-immunosuppressive doses of FK506 or by GPI1046, its anti-fibrotic activity is only realized when ARNT-ARNT homodimers form, as opposed to formation of ARNT-AHR or ARNT-HIF1α heterodimers. Mechanisms underlying ARNTs dimerization decision to specifically form ARNT-ARNT homodimers and possible cues to specifically induce ARNT homodimerization have been previously unknown. We here aimed to elucidate the molecular mechanisms underlying control of ARNT dimerization decision and to explore its therapeutic potential. Method We analyzed dimerization of recombinant and native ARNT by immunoprecipitation, MALDI-TOF mass spectrometry, and LS-MS/MS analysis and proximity ligation assay. Phosphorylation sites were mapped through generation of phosphorylation site mutants and through pharmacological inhibition. For in vivo analysis we challenged mice with model of unilateral ureter obstruction and carbon tetrachloride to induce fibrosis in kidney and liver. Results Here we report that inhibition of PP2A phosphatase activity increases intracellular accumulation of ARNT-ARNT homodimers. This effect is dependent on enhanced ARNT-ARNT homodimerization and decreased ARNT proteolytic degradation, but independent of ARNT transcription (which remains unchanged upon PP2A inhibition). We further identify that Ser77 phosphorylation plays a critical role in ARNT homodimerization, as ARNT-ARNT homodimers do not form with Ser77/Asp-mutant ARNT proteins. In light of previous studies which identified anti-fibrotic activity upon increased ARNT expression, we further demonstrate attenuated fibrosis upon monotherapy with the PP2A inhibitor LB100, and additive anti-fibrotic activities upon combination with pharmacological inducers of ARNT expression FK506 or GPI1046 in murine models of kidney and liver fibrosis. Conclusion Our study provides additional evidence for the anti-fibrotic activity of ARNT and reveals Ser77 phosphorylation as a novel pharmacological target to realize the therapeutic potential of increased ARNT transactivation activity.


Cancers ◽  
2018 ◽  
Vol 10 (10) ◽  
pp. 373 ◽  
Author(s):  
Arnatchai Maiuthed ◽  
Chuanpit Ninsontia ◽  
Katharina Erlenbach-Wuensch ◽  
Benardina Ndreshkjana ◽  
Julienne Muenzner ◽  
...  

The oncogenic cytoplasmic p21 contributes to cancer aggressiveness and chemotherapeutic failure. However, the molecular mechanisms remain obscure. Here, we show for the first time that cytoplasmic p21 mediates 5-Fluorouracil (5FU) resistance by shuttling p-Chk2 out of the nucleus to protect the tumor cells from its pro-apoptotic functions. We observed that cytoplasmic p21 levels were up-regulated in 5FU-resistant colorectal cancer cells in vitro and the in vivo Chorioallantoic membrane (CAM) model. Kinase array analysis revealed that p-Chk2 is a key target of cytoplasmic p21. Importantly, cytoplasmic form of p21 mediated by p21T145D transfection diminished p-Chk2-mediated activation of E2F1 and apoptosis induction. Co-immunoprecipitation, immunofluorescence, and proximity ligation assay showed that p21 forms a complex with p-Chk2 under 5FU exposure. Using in silico computer modeling, we suggest that the p21/p-Chk2 interaction hindered the nuclear localization signal of p-Chk2, and therefore, the complex is exported out of the nucleus. These findings unravel a novel mechanism regarding an oncogenic role of p21 in regulation of resistance to 5FU-based chemotherapy. We suggest a possible value of cytoplasmic p21 as a prognosis marker and a therapeutic target in colorectal cancer patients.


2021 ◽  
Vol 22 (4) ◽  
pp. 2037
Author(s):  
Bo-Yuan Hsiao ◽  
Chia-Hsin Chen ◽  
Ho-Yi Chi ◽  
Pei-Ru Yen ◽  
Ying-Zhen Yu ◽  
...  

Regulation of cellular actin dynamics is pivotal in driving cell motility. During cancer development, cells migrate to invade and spread; therefore, dysregulation of actin regulators is often associated with cancer progression. Here we report the role of ABRACL, a human homolog of the Dictyostelium actin regulator Costars, in migration and tumorigenic growth of cancer cells. We found a correlation between ABRACL expression and the migratory ability of cancer cells. Cell staining revealed the colocalization of ABRACL and F-actin signals at the leading edge of migrating cells. Analysis of the relative F-/G-actin contents in cells lacking or overexpressing ABRACL suggested that ABRACL promotes cellular actin distribution to the polymerized fraction. Physical interaction between ABRACL and cofilin was supported by immunofluorescence staining and proximity ligation. Additionally, ABRACL hindered cofilin-simulated pyrene F-actin fluorescence decay in vitro, indicating a functional interplay. Lastly, analysis on a colorectal cancer cohort demonstrated that high ABRACL expression was associated with distant metastasis, and further exploration showed that depletion of ABRACL expression in colon cancer cells resulted in reduced cell proliferation and tumorigenic growth. Together, results suggest that ABRACL modulates actin dynamics through its interaction with cofilin and thereby regulates cancer cell migration and participates in cancer pathogenesis.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Nora Bengoa-Vergniory ◽  
Elisavet Velentza-Almpani ◽  
Ana Maria Silva ◽  
Connor Scott ◽  
Mariana Vargas-Caballero ◽  
...  

Abstract Background Multimerization is a key process in prion-like disorders such as Alzheimer’s disease (AD), since it is a requirement for self-templating tau and beta-amyloid amyloidogenesis. AT8-immunohistochemistry for hyperphosphorylated tau is currently used for the diagnosis and staging of tau pathology. Given that tau–tau interactions can occur in the absence of hyperphosphorylation or other post-translational modifications (PTMs), the direct visualization of tau multimerization could uncover early pathological tau multimers. Methods Here, we used bimolecular fluorescent complementation, rapamycin-dependent FKBP/FRB-tau interaction and transmission electron microscopy to prove the in vitro specificity of tau-proximity ligation assay (tau-PLA). We then analyzed MAPT KO and P301S transgenic mice, and human hippocampus and temporal isocortex of all Braak stages with tau-PLA and compared it with immunohistochemistry for the diagnostic antibody AT8, the early phosphorylation-dependent AT180, and the conformational-dependent antibody MC1. Finally, we performed proteinase-K treatment to infer the content of amyloidogenic beta-sheet fold. Results Our novel tau-proximity ligation assay (tau-PLA) directly visualized tau–tau interactions in situ, and exclusively recognized tau multimers but not monomers. It elicited no signal in MAPT KO mouse brains, but extensively labelled P301S transgenic mice and AD brain. Two groups of structures were detected, a previously unreported widespread small-sized diffuse pathology and large, neurofibrillary-like lesions. Tau-PLA-labelled diffuse pathology appeared from the earliest Braak stages, mostly unaccompanied by tangle-like tau-immunohistochemistry, being significantly more sensitive than any small-sized dot-/thread-like pathology labelled by AT180-, AT8- and MC1-immunohistochemistry in most regions quantified at stages 0-II. Tau-PLA-labelled diffuse pathology was extremely sensitive to Proteinase-K, in contrast to large lesions. Conclusions Tau-PLA is the first method to directly visualize tau multimers both in vitro and in situ with high specificity. We find that tau multimerization appears extensively from the earliest presymptomatic Braak stages as a previously unreported type of diffuse pathology. Importantly, in our study multimerization is the earliest detectable molecular event of AD tau pathology. Our findings open a new window to the study of early tau pathology, with potential implications in early diagnosis and the design of therapeutic strategies.


Blood ◽  
2016 ◽  
Vol 127 (18) ◽  
pp. 2231-2240 ◽  
Author(s):  
Catherine Strassel ◽  
Nathalie Brouard ◽  
Lea Mallo ◽  
Nicolas Receveur ◽  
Pierre Mangin ◽  
...  

Key Points Emergence of a CD34+CD41low population with a high capacity to generate proplatelet-producing MKs and functional platelet-like elements. Platelet production is inversely correlated to CYP1B1 expression, a target of the aryl hydrocarbon receptor.


2019 ◽  
Author(s):  
Ηλιάννα Ζώη

Εισαγωγή: Ο human epidermal growth factor receptor 2 (HER-2) υπερεκφράζεται στο 20-30% των περιπτώσεων καρκίνου του μαστού (ΚΜ) και σχετίζεται με κακή πρόγνωση. Σηματοδοτικά μονοπάτια καθοδικά του υποδοχέα receptor activator of nuclear factor-κB (RANK) παίζουν κεντρικό ρόλο στον ΚΜ. Το σηματοδοτικό μονοπάτι RANKL/RANK εμπλέκεται στον HER-2(+) ΚΜ. Ο μοριακός μηχανισμός της συμμετοχής του στην καρκινογένεση παραμένει αδιερεύνητος. Σκοπός: Σκοπός της παρούσας διδακτορικής διατριβής είναι η διερεύνηση της εμπλοκής του RANK υποδοχέα, και του σηματοδοτικού μονοπατιού του NF-κB στην ογκογένεση των HER-2(+) καρκινωμάτων μαστού (ΚΜ). Επιπλέον στόχος είναι η αποσαφήνιση του μοριακού μηχανισμού ρύθμισης της RANKL/RANK σηματοδότησης από μέλη της οικογένειας EGFR μέσω πιθανής φυσικής αλληλεπίδρασης, επηρεάζοντας βασικές κυτταρικές ιδιότητες. Τελικός στόχο της μελέτης αποτελεί η in vitro αξιολόγηση της συνδυαστικής αναστολής των RANK και EGFR υποδοχέων στην εξέλιξη των καρκινικών κυττάρων γεγονός που συμβάλει σε νέες πιθανές θεραπευτικές προτάσεις. Υλικά και Μέθοδοι: Χρησιμοποιήθηκαν οι ανθρώπινες κυτταρικές σειρές ΚΜ, SKBR3, BT474, MCF7, MDA-MB-453. Μελετήθηκε η έκφραση των RANK και RANKL, μέσω RT-PCR, Western-Blot και τεχνικές ανοσοφθορισμού. Η αλληλεπίδραση του RANK με τον HER-2 ανιχνεύθηκε μέσω της μεθόδου Proximity Ligation Assay (PLA), η οποία επιτρέπει την οπτικοποίηση πρωτεϊνικών αλληλεπιδράσεων. Η ενεργοποίηση του NF-κB μονοπατιού μελετήθηκε μέσω Western-Blot. Χρησιμοποιήθηκαν οι αναστολείς trastuzumab (T), pertuzumab (P), denosumab (D) των δύο μελετώμενων μονοπατιών. Εφαρμόστηκαν λειτουργικές δοκιμασίες μετανάστευσης, απόπτωσης και πολλαπλασιασμού. Ο κυτταρικός πολλαπλασιασμός μελετήθηκε εκτιμώντας τη βιωσιμότητα των κυττάρων με τη δοκιμασία ΧTT και του clonogenic assay, το μεταναστευτικό δυναμικό με τη βιοδοκιμή προσομοίωσης τραύματος δι’ αμυχής (Scratch-Wound Healing Assay), ενώ η απόπτωση μέσω FACS έπειτα από χρώση των κυττάρων με ανεξίνη V. Αποτελέσματα: Η πρωτεϊνική έκφραση του υποδοχεα RANK και του συνδέτη του RANKL εντοπίστηκε σε όλες τις κυτταρικές σειρές. Ο υποδοχέας RANK βρέθηκε ότι διμερίζεται με τα μέλη της οικογένειας EGFR. Ο αριθμός των διμερών RANK/ HER-2 ανά κύτταρο φαίνεται να εξαρτάται από την έκφραση του HER-2 (SKBR3:5.4,BT474:8.2,MCF7:0.7,MDA-MB-453:0.3). Τα διμερή RANK/ HER-2 μειώνονται παρουσία των αναστολέων Denosumab (D), Trastuzumab (T) και Pertuzumab (P), ενώ αυξάνονται παρουσία του RANKL (R) στα κύτταρα SKBR3 (m:5.4, D:1.2, T:1.9, DT:0.6, TP:1, DTP:0.4, R:11.8) και BT474 (m:8.2, D:3.1, T:4.3, DT:0.7, TP:3.4, DTP:3.2, R:11.6). Η συνδυαστική επώαση των κυττάρων SKBR3 με τους αναστολείς μειώνει περαιτέρω την ενεργοποίηση του μονοπατιού NF-κB σε σχέση με τη μονή στόχευση. Η αναστολή των RANKL και HER-2 στη κυτταρική σειρά SKBR3 έχει ως αποτέλεσμα τον μειωμένο κυτταρικό πολλαπλασιασμό, αυξημένη απόπτωση και χαμηλότερο μεταναστευτικό δυναμικό συγκριτικά με τα κύτταρα ελέγχου (m) ενώ αντίθετες τιμές παρατηρήθηκαν παρουσία RANKL. Η συνδυαστική επώαση των κυττάρων SKBR3 με D, T και P πλεονεκτεί έναντι της μονής στόχευσης στις λειτουργικές δοκιμασίες. Το denosumab κατέστειλε την NF-κΒ σηματοδότηση και ελάττωσε το ρυθμό πολλαπλασιασμού στα καρκινικά κύτταρα MDA-MB-453. Η κυτταρική σειρά MCF7 δεν ανταποκρίθηκε στους αναστολείς. Συμπεράσματα: Τα αποτελέσματα μας καταδεικνύουν μια νέα φυσική και μοριακή συσχέτιση μεταξύ των HER-2 και RANK μονοπατιών, η οποία επηρεάζει την εξέλιξη των HER-2(+) ΚΜ. Επιπλέον, παρουσιάζονται ενδείξεις που καθιστούν τη συνδυαστική στόχευση των RANKL, HER-2 ως ωφέλιμη θεραπευτική στρατηγική, η οποία θα πρέπει να ελεγχθεί περαιτέρω σε συγκεκριμένους ασθενείς με ΚΜ.


BMC Cancer ◽  
2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Mehraj-U-Din Lone ◽  
Javed Miyan ◽  
Mohammad Asif ◽  
Showkat A. Malik ◽  
Parul Dubey ◽  
...  

Abstract Background The mechanistic (or mammalian) target of rapamycin (mTOR), a Ser/Thr kinase, associates with different subunits forming two functionally distinct complexes, mTORC1 and mTORC2, regulating a diverse set of cellular functions in response to growth factors, cellular energy levels, and nutrients. The mechanisms regulating mTORC1 activity are well characterized; regulation of mTORC2 activity, however, remains obscure. While studies conducted in Dictyostelium suggest a possible role of Ras protein as a potential upstream regulator of mTORC2, definitive studies delineating the underlying molecular mechanisms, particularly in mammalian cells, are still lacking. Methods Protein levels were measured by Western blotting and kinase activity of mTORC2 was analyzed by in vitro kinase assay. In situ Proximity ligation assay (PLA) and co-immunoprecipitation assay was performed to detect protein-protein interaction. Protein localization was investigated by immunofluorescence and subcellular fractionation while cellular function of mTORC2 was assessed by assaying extent of cell migration and invasion. Results Here, we present experimental evidence in support of the role of Ras activation as an upstream regulatory switch governing mTORC2 signaling in mammalian cancer cells. We report that active Ras through its interaction with mSIN1 accounts for mTORC2 activation, while disruption of this interaction by genetic means or via peptide-based competitive hindrance, impedes mTORC2 signaling. Conclusions Our study defines the regulatory role played by Ras during mTORC2 signaling in mammalian cells and highlights the importance of Ras-mSIN1 interaction in the assembly of functionally intact mTORC2.


2021 ◽  
Vol 5 (1) ◽  
pp. e202101115
Author(s):  
Yueyuan Yin ◽  
Fei Yan ◽  
Ruimin Zhou ◽  
Mingchen Li ◽  
Jinyi Ma ◽  
...  

Single-domain antibody (sdAb) holds the promising strategies for diverse research and translational applications. Here, we describe a method for the adaptation of the in situ proximity ligation assay (isPLA) followed by sequencing (isPLA-seq) to facilitate screening of a high-sensitive, high-throughput sdAb library for a given protein at subcellular and single-cell resolution. Based on the sequence of complementarity-determining region 3 (CDR3), the recombinant sdAb can be produced for in vitro and in vivo utilities. This method provides a general means to identify the functional measure of sdAb and its complementary epitopes and its potential applications to investigate cellular processes.


1996 ◽  
Vol 76 (05) ◽  
pp. 774-779 ◽  
Author(s):  
John T Brandt ◽  
Carmen J Julius ◽  
Jeanne M Osborne ◽  
Clark L Anderson

SummaryImmune-mediated platelet activation is emerging as an important pathogenic mechanism of thrombosis. In vitro studies have suggested two distinct pathways for immune-mediated platelet activation; one involving clustering of platelet FcyRIIa, the other involving platelet-associated complement activation. HLA-related antibodies have been shown to cause platelet aggregation, but the mechanism has not been clarified. We evaluated the mechanism of platelet aggregation induced by HLA-related antibodies from nine patients. Antibody to platelet FcyRIIa failed to block platelet aggregation with 8/9 samples, indicating that engagement of platelet FcyRIIa is not necessary for the platelet aggregation induced by HLA-related antibodies. In contrast, platelet aggregation was blocked by antibodies to human C8 (5/7) or C9 (7/7). F(ab’)2 fragments of patient IgG failed to induce platelet activation although they bound to HLA antigen on platelets. Intact patient IgG failed to aggregate washed platelets unless aged serum was added. The activating IgG could be adsorbed by incubation with lymphocytes and eluted from the lymphocytes. These results indicate that complement activation is involved in the aggregation response to HLA-related antibodies. This is the first demonstration of complement-mediated platelet aggregation by clinical samples. Five of the patients developed thrombocytopenia in relationship to blood transfusion and two patients developed acute thromboembolic disease, suggesting that these antibodies and the complement-dependent pathway of platelet aggregation may be of clinical significance.


2020 ◽  
Vol 17 ◽  
Author(s):  
Akhlesh Kumar Jain ◽  
Hitesh Sahu ◽  
Keerti Mishra ◽  
Suresh Thareja

Aim: To design D-Mannose conjugated 5-Fluorouracil (5-FU) loaded Jackfruit seed starch nanoparticles (JFSSNPs) for site specific delivery. Background: Liver cancer is the third leading cause of death in world and fifth most often diagnosed cancer is the major global threat to public health. Treatment of liver cancer with conventional method bears several side effects, thus to undertake these side effects as a formulation challenge, it is necessary to develop novel target specific drug delivery system for the effective and better localization of drug into the proximity of target with restricting the movement of drug in normal tissues. Objective: To optimize and characterize the developed D-Mannose conjugated 5-Fluorouracil (5-FU) loaded Jackfruit seed starch nanoparticles (JFSSNPs) for effective treatment of liver cancer. Materials and methods: 5-FU loaded JFSSNPs were prepared and optimized formulation had higher encapsulation efficiency were conjugated with D-Mannose. These formulations were characterized for size, morphology, zeta potential, X-Ray Diffraction, and Differential Scanning Calorimetry. Potential of NPs were studied using in vitro cytotoxicity assay, in vivo kinetic studies and bio-distribution studies. Result and discussion: 5-Fluorouracil loaded NPs had particle size between 336 to 802nm with drug entrapment efficiency was between 64.2 to 82.3%. In XRD analysis, 5-FU peak was diminished in the diffractogram, which could be attributed to the successful incorporation of drug in amorphous form. DSC study suggests there was no physical interaction between 5- FU and Polymer. NPs showed sustained in vitro 5-FU release up to 2 hours. In vivo, mannose conjugated NPs prolonged the plasma level of 5-FU and assist selective accumulation of 5-FU in the liver (vs other organs spleen, kidney, lungs and heart) compared to unconjugated one and plain drug. Conclusion: In vivo, bio-distribution and plasma profile studies resulted in significantly higher concentration of 5- Fluorouracil liver suggesting that these carriers are efficient, viable, and targeted carrier of 5-FU treatment of liver cancer.


Sign in / Sign up

Export Citation Format

Share Document