[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (29)

Search Parameters:
Keywords = legumain

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 4416 KiB  
Article
Cancer Cell Secreted Legumain Promotes Gastric Cancer Resistance to Anti-PD-1 Immunotherapy by Enhancing Macrophage M2 Polarization
by Xu Pei, Shi-Long Zhang, Bai-Quan Qiu, Peng-Fei Zhang, Tian-Shu Liu and Yan Wang
Pharmaceuticals 2024, 17(7), 951; https://doi.org/10.3390/ph17070951 - 16 Jul 2024
Viewed by 753
Abstract
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in [...] Read more.
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in modulating the tumor immune microenvironment and the biological effect on the immune evasion of gastric cancer remains unclear. In this study, we found that forced expression of sLGMN in gastric cancer serum correlates with increased M2 macrophage infiltration in GC tissues and predicted resistance to anti-PD-1 immunotherapy. Mechanistically, gastric cancer cells secrete LGMN via binding to cell surface Integrin αvβ3, then activate Integrin αvβ3/PI3K (Phosphatidylinositol-4,5-bisphosphate3-kinase)/AKT (serine/threonine kinase)/mTORC2 (mammalian target of rapamycin complex 2) signaling, promote metabolic reprogramming, and polarize macrophages from the M1 to the M2 phenotype. Either blocking LGMN, Integrin αv, or knocking out Integrin αv expression and abolishing the LGMN/Integrin αvβ3 interaction significantly inhibits metabolic reprogramming and polarizes macrophages from the M1 to the M2 phenotype. This study reveals a critical molecular crosstalk between gastric cancer cells and macrophages through the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis in promoting gastric cancer immune evasion and resistance to anti-PD-1 immunotherapy, indicating that the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis may act as a promising therapeutic target. Full article
(This article belongs to the Special Issue Small Molecules in Cancer Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Relationship between LGMN expression levels and M2 macrophage infiltration in gastric cancer. (<b>A</b>) Analysis of TCGA database suggests a correlation between LGMN expression levels in gastric cancer and M2 macrophage polarization. (<b>B</b>) Differential serum LGMN expression in patients sensitive and resistant to anti-PD-1 therapy. (<b>C</b>,<b>D</b>) Immunohistochemical detection of M1 or M2 macrophage infiltration in gastric cancer tissues, and their correlation with serum LGMN expression levels in gastric cancer patients (bar 100 μm). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>LGMN induces polarization of macrophages from M1 to M2 phenotype. (<b>A</b>) Co-culture of THP-1-derived M1 macrophages with ASG-L and BGC823-L cells followed by flow cytometry to detect expression of M2 macrophage markers CD163 and CD206, and the polarization of M1 macrophages towards M2 macrophages can be blocked by LGMN-neutralizing antibodies. (<b>B</b>) Incubation of THP-1-derived M1 macrophages with rh-LGMN protein for 5 days followed by flow cytometry to detect expression of M2 macrophage markers CD163 and CD206. (<b>C</b>) Stimulation of healthy human PBMCs with IFN-γ followed by flow cytometry to detect expression of CD80 and CD86. (<b>D</b>) Induction of M1 macrophages from PBMCs by culture with rh-LGMN, followed by flow cytometry to detect expression of CD206 and CD163.</p>
Full article ">Figure 3
<p>LGMN induces polarization of macrophages from M1 to M2 phenotype by forming a complex with Integrin αvβ3. (<b>A</b>) Western blot analysis of the effect of rh-LGMN, rh-LGMN + Integrin αv antibody, and rh-LGMN + Integrin β3 antibody on mTORC2 activity in M1 macrophages. (<b>B</b>) Flow cytometry analysis of the effect of rh-LGMN, rh-LGMN + Integrin αv antibody, and rh-LGMN + Integrin β3 antibody on the polarization of macrophages from M1 to M2 phenotype. (<b>C</b>) Establishment of M1 macrophage cells derived from THP-1 cells with knockdown of Integrin β3 using lentivirus. (<b>D</b>) Flow cytometry analysis of the effect of knocking down Integrin β3 expression on LGMN-mediated polarize macrophages from M1 to M2 phenotype. (<b>E</b>) Stimulation of M1 macrophages derived from PBMCs with rh-LGMN protein for 48 h followed by Western blot analysis to detect changes in mTORC2 signaling pathway activity. (<b>F</b>) Stimulation of M1 macrophages derived from healthy human PBMCs with rh-LGMN for 48 h followed by flow cytometry to detect changes in CD36 expression.</p>
Full article ">Figure 4
<p>mTOR inhibitor blocks LGMN-induced polarization of macrophages from M1 to M2 phenotype. (<b>A</b>) Flow cytometry analysis of the effect of AZD2014 on LGMN-mediated polarization of macrophages from M1 to M2 phenotype. (<b>B</b>) Western blot analysis of the effect of AZD2014 on upregulation of mTORC2 signaling pathway activity by rh-LGMN in M1 macrophages. (<b>C</b>) Western blot analysis of the effect of rh-LGMN on mTORC1 signaling pathway activity in M1 macrophages.</p>
Full article ">Figure 5
<p>rh-LGMN protein promotes glycolysis and fatty acid oxidation in M1 macrophages derived from healthy human PBMCs. (<b>A</b>) ECAR assay. (<b>B</b>) Fatty acid oxidation assay. (<b>C</b>) Oxygen consumption assay. (<b>D</b>) Fatty acid uptake assay. (<b>E</b>,<b>F</b>) Low-dose mTOR inhibitor AZD2014 inhibits rh-LGMN-induced glycolysis and oxidative phosphorylation in M1 macrophages. (<b>G</b>,<b>H</b>) Blocking antibodies against Integrin αv but not Integrin β3 inhibits rh-LGMN protein-induced glycolysis (<b>G</b>) and oxygen consumption (<b>H</b>) in M1 macrophages derived from healthy human PBMCs. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 ((<b>G</b>,<b>H</b>): LGMN vs. LGMN + Integrin αv).</p>
Full article ">Figure 6
<p>sLGMN induces resistance of gastric cancer to anti-PD-1 therapy. (<b>A</b>) Relationship between serum LGMN levels and PFS of gastric cancer to anti-PD-1 therapy. (<b>B</b>) Establishment of 615 mouse gastric cancer model using MCF cells, followed by different therapies to observe the relationship between sLGMN and sensitivity of gastric cancer to PD-1 mAb. (<b>C</b>) Statistical analysis of tumor volume after different treatments in 615 mouse MFC cell gastric cancer model. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; NS, not significant.</p>
Full article ">
22 pages, 3532 KiB  
Review
The Role of Cytokines and Molecular Pathways in Lung Fibrosis Following SARS-CoV-2 Infection: A Physiopathologic (Re)view
by Mihai Lazar, Mihai Sandulescu, Ecaterina Constanta Barbu, Cristina Emilia Chitu-Tisu, Darie Ioan Andreescu, Andreea Nicoleta Anton, Teodora Maria Erculescu, Alexandru Mihai Petre, George Theodor Duca, Vladimir Simion, Isabela Felicia Padiu, Cosmina Georgiana Pacurar, Ruxandra Rosca, Teodor Mihai Simian, Constantin Adrian Oprea and Daniela Adriana Ion
Biomedicines 2024, 12(3), 639; https://doi.org/10.3390/biomedicines12030639 - 13 Mar 2024
Cited by 1 | Viewed by 2245
Abstract
SARS-CoV-2 infection is a significant health concern that needs to be addressed not only during the initial phase of infection but also after hospitalization. This is the consequence of the various pathologies associated with long COVID-19, which are still being studied and researched. [...] Read more.
SARS-CoV-2 infection is a significant health concern that needs to be addressed not only during the initial phase of infection but also after hospitalization. This is the consequence of the various pathologies associated with long COVID-19, which are still being studied and researched. Lung fibrosis is an important complication after COVID-19, found in up to 71% of patients after discharge. Our research is based on scientific articles indexed in PubMed; in the selection process, we used the following keywords: “lung fibrosis”, “fibrosis mediators”, “fibrosis predictors”, “COVID-19”, “SARS-CoV-2 infection”, and “long COVID-19”. In this narrative review, we aimed to discuss the current understanding of the mechanisms of initiation and progression of post-COVID-19 lung fibrosis (PC-19-LF) and the risk factors for its occurrence. The pathogenesis of pulmonary fibrosis involves various mediators such as TGF-β, legumain, osteopontin, IL-4, IL-6, IL-13, IL-17, TNF-α, Gal-1, Gal-3, PDGF, and FGFR-1. The key cellular effectors involved in COVID-19 lung fibrosis are macrophages, epithelial alveolar cells, neutrophils, and fibroblasts. The main fibrosis pathways in SARS-CoV-2 infection include hypoxemia-induced fibrosis, macrophage-induced fibrosis, and viral-fibroblast interaction-induced fibrosis. Full article
(This article belongs to the Special Issue The Role of Cytokines in Health and Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>The flowchart of the review manuscript. Abbreviations:<sup>1</sup> from 2020 till 2023; <sup>2</sup> in case of unclear or contradictory information, the supervisors of our review (ML and DAI) were consulted.</p>
Full article ">Figure 2
<p>Fibroblast–cytokine interactions in SARS-CoV-2 infection. Abbreviations: Act1–adaptor protein Act1, AKT—alpha serine/threonine-protein kinase, EBPs—enhancer-binding proteins, ECM—extracellular matrix, ERK—extracellular signal-regulated protein kinase, FAK—focal adhesion kinase 1, FN—fibronectine, IKK—IkappaB kinase, IL—interleukin, IL6-R—receptor for interleukin 6, IL17-R—receptor for interleukin 17, IL11-R—receptor for interleukin 11, Gal—galectin, JAK/STAT—Janus kinase/signal transducer and activator of transcription, MAPK/MEK—mitogen-activated protein kinases, mTOR—<span class="html-italic">mechanistic target of rapamycin</span>, NF-κB—nuclear factor kappa-light-chain-enhancer, PDGF—platelet derived growth factor, PI3K—phosphoinositide 3-kinase, RAF—rapidly accelerated fibrosarcoma kinase, SHP2—Src homology 2 domain-containing <span class="html-italic">protein tyrosine phosphatase,</span> α-SMA—α smooth muscle actin, SMAD—mothers against decapentaplegic homolog 1, STAT3—<span class="html-italic">signal transducer and activator of transcription 3</span>, TAK—transforming growth factor beta activated kinase, TGF-β—transforming growth factor beta, TLR—Toll-like receptor, TNF α—tumor necrosis factor α, tumor necrosis factor α receptor—TNFR, TRAF—tumor necrosis factor receptor-associated factor, and TYK-tyrosine kinase.</p>
Full article ">Figure 3
<p>Fibrosis pathways in SARS-CoV-2 infection. Steps to lung fibrosis: S1. alveolar injury; S2. macrophages and neutrophils activation; S3. hypoxemia; S4. upregulation of HIF-1α and profibrotic genes; S5. production of inflammatory cytokines; S6. fibroblast–cytokine/SARS-CoV-2 interactions; S7. fibroblast migration, proliferation, and differentiation; S8. ECM changes (lung fibrosis). Abbreviations: AEC I—alveolar epithelial cell type I, AECs type II—alveolar epithelial cells type II, ECM—extracellular matrix, FAK 1—focal adhesion kinase 1, IL—interleukin, HIF-1α—hypoxia-inducible factor-1α, IL6-R—receptor for interleukin 6, IL17-R—receptor for interleukin 17, Gal—galectin, JAK/STAT—Janus kinase/signal transducer and activator of transcription, PDGF—platelet-derived growth factor, PAI—plasminogen activator inhibitor-1, MAMPS—microbe-associated molecular patterns, NF-κB—nuclear factor kappa-light-chain-enhancer, SMAD—mothers against decapentaplegic homolog 1, STAT3—signal transducer and activator of transcription 3, TGF-β—transforming growth factor beta, TLR—Toll-like receptor, TREM2—triggering receptor expressed on myeloid cells 2, TNF α—tumor necrosis factor α, tumor necrosis factor α receptor—TNFR, and α-SMA—α smooth muscle actin.</p>
Full article ">
23 pages, 5952 KiB  
Article
UHPLC-MS/MS Assay for Quantification of Legubicin, a Novel Doxorubicin-Based Legumain-Activated Prodrug, and Its Application to Pharmacokinetic and Tissue Distribution Studies
by Liyuan Ma, Qiaoling Yu, Meng Zhuang, Chen Yang, Yuan Liu, Yuling Li, Cheng Liu, Xiaoyan Shen and Yan Chang
Molecules 2024, 29(4), 775; https://doi.org/10.3390/molecules29040775 - 8 Feb 2024
Cited by 1 | Viewed by 1176
Abstract
Legubicin, a novel prodrug based on doxorubicin, has both albumin-binding and legumain-activating properties. The aim of this study was to develop and validate a UHPLC-MS/MS method for investigating the in vivo pharmacokinetics and tissue distribution profiles of legubicin in rats and tumor-bearing mice [...] Read more.
Legubicin, a novel prodrug based on doxorubicin, has both albumin-binding and legumain-activating properties. The aim of this study was to develop and validate a UHPLC-MS/MS method for investigating the in vivo pharmacokinetics and tissue distribution profiles of legubicin in rats and tumor-bearing mice following intravenous administration, and to compare this prodrug with the positive control drug doxorubicin. The study employed a UHLC-MS/MS method to determine the levels of albumin-bound of legubicin and two metabolites (free Leu-DOX and DOX) in plasma, tumor, and tissue samples. This method was validated for good selectivity, high sensitivity, excellent extraction recovery, and short run time. The results showed that legubicin was present in the circulation in vivo mainly in a protein-bound form with larger AUC values and lower clearance and distribution, and essentially released small amounts of doxorubicin. Compared to administration of equimolar doses of doxorubicin, legubicin showed increased exposure of the active drug in the tumor and decreased the level of the active drug in the heart and kidney. This study provides valuable information on the pharmacokinetics and tissue distribution of legubicin, implicating its potential as a novel and effective drug candidate for anti-cancer therapies. Full article
(This article belongs to the Section Analytical Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structures of Legubicin (<b>a</b>), Leu-DOX (<b>b</b>), and DOX (<b>c</b>).</p>
Full article ">Figure 1 Cont.
<p>Chemical structures of Legubicin (<b>a</b>), Leu-DOX (<b>b</b>), and DOX (<b>c</b>).</p>
Full article ">Figure 2
<p>(<b>a</b>) Incubation kinetics of legubicin with HSA at room temperature (<span class="html-italic">n</span> = 1); (<b>b</b>) legubicin’s plasma stability profile (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 3
<p>(<b>a</b>) Chromatograms of Dox (red); Leu-Dox (green); legubicin–HSA conjugate (powder blue and ink blue); and legubicin–HSA conjugate incubated with legumain for 1 h (blue), 6 h (black), and 20 h (pink). The specific liquid-phase conditions are shown as follows. Column: Agilent Eclipse Plus 4.6 × 250 mm, 5 µm; mobile phase A: 0.1% trifluoroacetic acid in water; mobile phase B: acetonitrile. Flow rate: 1.0 mL/min; HPLC: Waters 2695; detector: Waters 2475 detector (ex470/em600); column temperature: 30 °C; injection volume: 10 µL; run time: 30 min; needle wash solvent: 50% acetonitrile in water; gradient elution: starting 20% B, increasing from 20% to 45% B in 15 min, increasing from 45% to 80% B in 15 to 20 min, maintained for 5 min, then decreasing to 20% B in 0.1 min, and continuing until the end of 30 min. (<b>b</b>) Kinetics of Leu-Dox release from legubicin (black) and legubicin–albumin conjugate (blue) in the presence of legumain (<span class="html-italic">n</span> = 4). (<b>c</b>) Stability of Leu-Dox in this enzymatic system (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>Profiles of mean plasma concentration versus time of (<b>a</b>) Leu-DOX (female), (<b>b</b>) Leu-DOX (male), (<b>c</b>) DOX (female), (<b>d</b>) DOX (male), and (<b>e</b>) albumin conjugate of legubicin, after single dose administration (1.5, 5, and 15 mg/kg, respectively) in Sprague-Dawley rats (<b>a</b>–<b>d</b>, <span class="html-italic">n</span> = 3; <b>e</b>, <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 5
<p>Profiles of mean plasma concentration versus time of (<b>a</b>,<b>b</b>) Leu-DOX (Female), (<b>c</b>,<b>d</b>) Leu-DOX (Male), (<b>e</b>,<b>f</b>) DOX (Female), (<b>g</b>,<b>h</b>) DOX (Male), and (<b>i</b>,<b>j</b>) albumin conjugate of legubicin. At left, the comparison between days 1 and 22, and at right, the continuous weekly dosing of 1.5 mg/kg of legubicin for four weeks in Sprague-Dawley rats ((<b>a</b>–<b>h</b>), <span class="html-italic">n</span> = 3; (<b>i</b>,<b>j</b>), <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 6
<p>Mean tissue–drug/plasma–drug concentrations in tissues and plasma of tumor-bearing mice after intravenous administration of legubicin of (<b>a</b>) Leu-DOX; (<b>b</b>) DOX, or administration of doxorubicin (<b>c</b>) (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 7
<p>Comparison of doxorubicin exposure (AUC<sub>last</sub>) in plasma, heart, kidney, and tumor, after intravenous administration of doxorubicin or legubicin, in tumor-bearing mice (<span class="html-italic">n</span> = 6).</p>
Full article ">
25 pages, 6852 KiB  
Article
Trichomonas vaginalis Legumain-2, TvLEGU-2, Is an Immunogenic Cysteine Peptidase Expressed during Trichomonal Infection
by Esly Alejandra Euceda-Padilla, Miriam Guadalupe Mateo-Cruz, Leticia Ávila-González, Claudia Ivonne Flores-Pucheta, Jaime Ortega-López, Daniel Talamás-Lara, Beatriz Velazquez-Valassi, Lidia Jasso-Villazul and Rossana Arroyo
Pathogens 2024, 13(2), 119; https://doi.org/10.3390/pathogens13020119 - 27 Jan 2024
Viewed by 1660
Abstract
Trichomonas vaginalis is the causative agent of trichomoniasis, the most prevalent nonviral, neglected sexually transmitted disease worldwide. T. vaginalis has one of the largest degradomes among unicellular parasites. Cysteine peptidases (CPs) are the most abundant peptidases, constituting 50% of the degradome. Some CPs [...] Read more.
Trichomonas vaginalis is the causative agent of trichomoniasis, the most prevalent nonviral, neglected sexually transmitted disease worldwide. T. vaginalis has one of the largest degradomes among unicellular parasites. Cysteine peptidases (CPs) are the most abundant peptidases, constituting 50% of the degradome. Some CPs are virulence factors recognized by antibodies in trichomoniasis patient sera, and a few are found in vaginal secretions that show fluctuations in glucose concentrations during infection. The CPs of clan CD in T. vaginalis include 10 genes encoding legumain-like peptidases of the C13 family. TvLEGU-2 is one of them and has been identified in multiple proteomes, including the immunoproteome obtained with Tv (+) patient sera. Thus, our goals were to assess the effect of glucose on TvLEGU-2 expression, localization, and in vitro secretion and determine whether TvLEGU-2 is expressed during trichomonal infection. We performed qRT-PCR assays using parasites grown under different glucose conditions. We also generated a specific anti-TvLEGU-2 antibody against a synthetic peptide of the most divergent region of this CP and used it in Western blot (WB) and immunolocalization assays. Additionally, we cloned and expressed the tvlegu-2 gene (TVAG_385340), purified the recombinant TvLEGU-2 protein, and used it as an antigen for immunogenicity assays to test human sera from patients with vaginitis. Our results show that glucose does not affect tvlegu-2 expression but does affect localization in different parasite organelles, such as the plasma membrane, Golgi complex, hydrogenosomes, lysosomes, and secretion vesicles. TvLEGU-2 is secreted in vitro, is present in vaginal secretions, and is immunogenic in sera from Tv (+) patients, suggesting its relevance during trichomonal infection. Full article
(This article belongs to the Special Issue Trichomonas vaginalis Infection)
Show Figures

Figure 1

Figure 1
<p>In silico analysis of the <span class="html-italic">tvlegu-2</span> gene and amino acid sequences. (<b>A</b>) Diagram of the <span class="html-italic">tvlegu-2</span> gene (TVAG_385340) in contig DS114117. Red: <span class="html-italic">tvlegu-2</span> gene with a size of 1179 bp; light blue: flanking genes encoding a putative thioredoxin (TVAG_385350) and a putative RNA-binding protein (TVAG_385330). The 5′ intergenic region is 4575 nt. (<b>Aa</b>) The 5′ regulatory region was analyzed and contains an Inr consensus sequence for transcription initiation (underlined); +1 indicates the possible transcription start site (black bend arrow). (<b>Ab</b>) The 3′ intergenic region is 718 nt. The 3′ downstream region contains two probable polyadenylation sites (PS<sub>1</sub> and PS<sub>2</sub>, double underlining), two probable cleavage sites (CS<sub>1</sub> and CS<sub>2</sub>; black arrow), and two downstream element sequences (DSE<sub>1</sub> and DSE<sub>2</sub>; triple underline). (<b>B</b>) Structure and putative posttranslational modifications of the 392 aa precursor TvLEGU-2 protein. Pink: signal peptide (SP), green: catalytic domain (CD), blue: activation peptide (AP), and yellow: C-terminal domain (LSAM). (<b>Ba</b>) Yellow circles correspond to predicted phosphorylation sites; pink hexagon corresponds to a putative glycosylation site with a score &gt;0.7; TRAF6 motif (blue star, 32 picks); catalytic residues: Asn (N)/His (H)/Cys (<b>c</b>) (black star); cleavage sites (black arrows). (<b>Bb</b>) Theoretical three-dimensional (3-D) model of TvLEGU-2 in AlphaFold2 v1.5.2. The 3-D model of TvLEGU-2 pro-peptidase, location of the His/Cys (His-Gly-X-Ala-Cys) catalytic dyad (close-up). (<b>Bc</b>) Putative processing steps of TvLEGU-2. Inactive proenzyme is synthesized at a neutral pH and undergoes autocatalytic processing at the C-terminal Asn<sup>301</sup> (pH &lt; 5.5) and K<sup>270</sup> (pH ≤ 4.0) sites and N-terminally after Cys<sup>11</sup>. The N-terminal processing is not essential for enzyme activation, but the release of the C-terminal LSAM domain is essential to gaining full AEP activity.</p>
Full article ">Figure 2
<p>Effect of glucose on the mRNA and protein expression of TvLEGU-2. (<b>A</b>) qPCR with specific primers for the <span class="html-italic">tvlegu-2</span> and <span class="html-italic">α-tubulin</span> genes using 100 ng of cDNA from parasites grown in glucose restriction (GR) and high-glucose (HG) conditions. <span class="html-italic">α-tubulin</span> was used as a normalizing gene; ns, no significant differences. (<b>B</b>) Western blot assay of PREs from parasites grown under GR (&lt;1 mM) and HG (50 mM). Coomassie Brilliant-Blue (CBB)-stained 10% SDS PAGE gel for PREs from parasites grown under GR and HG conditions (Lanes 2 and 3), respectively. For WB assays, duplicate gels were transferred onto NC membranes and incubated with different antibodies: Rα-TvLEGU-2pep (1:500 dilution) to detect the TvLEGU-2 protein, Rα-TvCP2r (1:6000 dilution) to detect a control protein overexpressed under GR conditions, Rα-TvTIM (1:1000 dilution) to detect an overexpressed TvTIM control protein under HG conditions, and a negative control with PI serum or no primary antibody (−). Arrowheads show the position of the native TvLEGU-2 (∼55, ∼49, ∼34, and ∼29 kDa) protein bands. (<b>C</b>) Silver-stained 2-DE protease-rich extracts from parasites grown in normal glucose conditions (25 mM) (<b>Ca</b>). WB of duplicate gels transferred onto NC membranes incubated with Rα-TvLEGU-1r (1:1000 dilution) antibody (<b>Cc</b>) to serve as a control for the specificity of Rα-TvLEGU-2pep antibody against other legumain proteins, Rα-TvLEGU-2pep (1:100 dilution) (<b>Cd</b>) to detect the TvLEGU-2 protein in PREs, or a negative control with PI serum or no primary antibody (−) (<b>Cb</b>). Arrowheads show the position of the native TvLEGU-2 (∼34 and ∼29 kDa) proteins. (<b>D</b>) Coomassie Brilliant Blue-stained, purified recombinant TvLEGU-1r protein (CBB; Lane 1). WB assays of TvLEGU-1r incubated with Rα-TvLEGU-1r (1:3000 dilution) (Lane 3), or Rα-TvLEGU-2pep (1:1000 dilution) (Lane 4) antibody or PI serum or only the secondary antibody as a negative control (−) (Lane 2). Arrowhead points to the recombinant protein band TvLEGU-1r (∼46 kDa). (<b>E</b>) Coomassie Brilliant Blue-stained, purified recombinant TvLEGU-2r protein (CBB; Lane 1). WB assays of TvLEGU-2r incubated with Rα-TvLEGU-2pep (1:1000 dilution) (Lane 4), or Rα-TvLEGU-1r (1:3000 dilution) (Lane 3) antibody, or PI serum or only the secondary antibody as a negative control (−) (Lane 2). Arrowhead points to the recombinant protein band TvLEGU-2r (∼85 kDa). kDa, molecular weight markers in kilodaltons (Bio-Rad).</p>
Full article ">Figure 3
<p>Localization of TvLEGU-2 in <span class="html-italic">T. vaginalis</span> under glucose restriction (GR) and high-glucose (HG) conditions. (<b>A</b>) IFA of nonpermeabilized parasites under GR (<b>Af</b>–<b>Aj</b>) and HG (<b>Ak</b>–<b>Ao</b>) conditions with Mα-TvLEGU-2pep (1:100 dilution) and Rα-Membrane (1:200 dilution) antibodies. Negative control with no primary antibody (−) (<b>Aa</b>–<b>Ae</b>). TvLEGU-2 (Alexa 647, red), membrane (FITC, green), and nucleus (DAPI, blue). White bar = 10 μm, white arrowheads = membrane colocalization, and white stars = vesicles. (<b>B</b>) IFA of permeabilized parasites under GR (<b>Bf</b>–<b>Bj</b>) and HG (<b>Bk</b>–<b>Bo</b>) conditions with Mα-TvLEGU-2pep (1:100 dilution) and Rα-Membrane (1:200 dilution) antibodies. The negative control with PI serum or no primary antibody (<b>Ba</b>–<b>Be</b>). TvLEGU-2 (Alexa 647, red), membrane (FITC, green), and nucleus (DAPI, blue). White bar = 10 μm, white stars = vesicles, white diamond = Golgi apparatus. (<b>C</b>) TEM under GR and HG conditions with Mα-TvLEGU-2pep antibody (1:10 dilution) with 30 nm gold particles. Negative control with PI serum or no primary antibody (−). Cytoplasm (C), hydrogenosome (H), membrane (M), nucleus (N), vesicle (V), autophagic vesicle (AV), endoplasmic reticulum (ER), and Golgi apparatus (G). Localization of TvLEGU-2: white arrowheads s = membrane, black arrows = extracellular, black stars = vesicles, black diamonds = Golgi apparatus, and black arrowheads = hydrogenosomes. Scale bar = 500 nm (<b>Cb</b>,<b>Cc</b>,<b>Cd</b>,<b>Cg</b>) and scale bar = 1000 nm (<b>Ca</b>,<b>Ce</b>,<b>Cf</b>,<b>Ch</b>).</p>
Full article ">Figure 4
<p>Colocalization of TvLEGU-2 and PFO in <span class="html-italic">T. vaginalis</span> hydrogenosomes under glucose restriction (GR) and high-glucose (HG) conditions. (<b>A</b>) IFA of permeabilized parasites under GR (<b>Af</b>–<b>Aj</b>) and HG (<b>Ak</b>–<b>Ao</b>) conditions with Mα-TvLEGU-2pep (1:100 dilution) and Rα-PFO50 (1:50) antibodies to label hydrogenosomes. Negative control with no primary antibody (−) (<b>Aa</b>–<b>Ae</b>). TvLEGU-2 (Alexa 647, red), nucleus (DAPI, blue), and PFO (FITC, green). White bar = 10 μm, and the white arrowheads indicate colocalization. (<b>B</b>) TEM analysis under GR and HG conditions with Mα-TvLEGU-2pep antibody (1:10 dilution) with 30 nm gold particles and Rα-PFO50 antibody (1:10 dilution) with 15 nm gold particles. Negative control with PI serum or no primary antibody (−). Cytoplasm (C), hydrogenosome (H), membrane (M), and vesicle (V). The black arrowheads indicate the close proximity of both labels, and the black stars indicate the localization of both proteins inside hydrogenosomes. Scale bar = 500 nm (<b>Ba</b>–<b>Bd</b>), and scale bar = 1000 nm (<b>Be</b>–<b>Bg</b>).</p>
Full article ">Figure 5
<p>Colocalization of TvLEGU-2 and TvAtg8b in <span class="html-italic">T. vaginalis</span> under glucose restriction (GR) and high-glucose (HG) conditions. (<b>A</b>) IFA of permeabilized parasites under GR (<b>Af</b>–<b>Aj</b>) and HG (<b>Ak</b>–<b>Ao</b>) conditions with Mα-TvLEGU-2pep (1:100) and Rα-rTvATG8b (1:50) antibodies to label autophagosomes. Negative control with PI serum or no primary antibody (−) (<b>Aa</b>–<b>Ae</b>). TvLEGU-2 (Alexa 647, red), TvAtg8b (FITC, green), and nucleus (DAPI, blue), white bar = 10 μm. The white arrowheads indicate colocalization. (<b>B</b>) TEM of parasites grown under GR and HG conditions with Mα-TvLEGU-2pep antibody (1:10 dilution) with 30 nm gold particles and Rα-rTvAtg8b (1:10 dilution) with 15 nm gold particles. Negative control with PI serum or no primary antibody (−). Cytoplasm (C), hydrogenosome (H), nucleus (N), vesicle (V), autophagic vesicle (AV), endoplasmic reticulum (ER), and Golgi apparatus (G). Black arrowhead = TvLEGU-2 and TvAtg8b localization in the same vesicle; black arrow = TvLEGU-2 and TvAtg8b localization in the same hydrogenosome. Scale bar = 500 nm (<b>Bb</b>,<b>Bd</b>,<b>Bg</b>), and scale bar = 1000 nm (<b>Ba</b>,<b>Bc</b>,<b>Be</b>,<b>Bf</b>).</p>
Full article ">Figure 6
<p>Colocalization of TvLEGU-2 and LysoTracker in <span class="html-italic">T. vaginalis</span> under glucose restriction (GR) and high-glucose (HG) conditions. IFA of permeabilized parasites under GR (<b>g</b>–<b>l</b>) and HG (<b>m</b>–<b>r</b>) conditions with Mα-TvLEGU-2pep (1:100) and LysoTracker 5 μM to label lysosomes. Negative control with PI serum or no primary antibody (−) (<b>a</b>–<b>f</b>). TvLEGU-2 (Alexa 647, red), LysoTracker (green), and nucleus (DAPI, blue). White bar = 10 μm. White arrowheads = colocalization of TvLEGU-2 and LysoTracker.</p>
Full article ">Figure 7
<p>Colocalization of TvLEGU-1 and TvLEGU-2 in <span class="html-italic">T. vaginalis</span> under glucose restriction (GR) and high-glucose (HG) conditions. IFA of permeabilized parasites under GR (<b>f</b>–<b>o</b>) and HG (<b>p</b>–<b>y</b>) conditions with Mα-TvLEGU-2pep (1:100) and Rα-TvLEGU-1r (1:300) antibodies. Negative control with PI serum or no primary antibody (−) (<b>a</b>–<b>e</b>). TvLEGU-1 (FITC, green), TvLEGU-2 (Alexa 647, red), and nucleus (DAPI, blue). White bar = 10 μm. White circles = membrane, white diamonds = Golgi apparatus, and white stars = vesicles.</p>
Full article ">Figure 8
<p>Glucose conditions differentially regulate zymogen and mature peptidase in vitro secretion of TvLEGU-2 by <span class="html-italic">Trichomonas vaginalis</span>. (<b>A</b>) PREs from parasites grown under GR (&lt;1 mM) and HG (50 mM) conditions were separated on 10% SDS PAGE gels, transferred onto NC membranes, and stained with Ponceau red (PR)- (Lanes 2 and 3: GR and HG, respectively). For WB assays, NC membranes were incubated with different antibodies: Rα-TvLEGU-2pep (1:500 dilution) to detect the TvLEGU-2 protein (Lanes 6 and 7) and PI serum or no primary antibody (−) to serve as a negative control (Lanes 4 and 5). Arrowheads show the position of the native TvLEGU-2 isoform (∼55, ∼34, and ∼29 kDa) proteins. (<b>B</b>) Western blot assay of SCPs from parasites grown under GR (&lt;1 mM) and HG (50 mM) conditions that were precipitated by TCA and separated on 10% SDS PAGE gels, transferred onto NC membranes, and PR-stained (Lanes 1 and 2: GR and HG, respectively). For WB assays, NC membranes were incubated with different antibodies: Rα-TvLEGU-2pep (1:500 dilution) to detect the TvLEGU-2 isoforms (Lanes 5 and 6) and PI serum or no primary antibody (−) to serve as a negative control (Lanes 3 and 4). Arrowheads show the positions of the native TvLEGU-2 isoform (∼55 and ∼29 kDa) proteins. kDa, molecular weight markers in kilodaltons (Bio-Rad).</p>
Full article ">Figure 9
<p>Presence of TvLEGU-2 during trichomonal infection. (<b>A</b>) Western blot assay of TvLEGU-2r incubated with Tv (+) and Tv (−) patient sera. Controls of the WB assay using the recombinant TvLEGU-2 Coomassie Brilliant Blue (CBB)-stained 10% SDS PAGE gel for TvLEGU-2r (Lane 1) and recognition by the anti-TvLEGU-2r antibody (1:10,000 dilution; Lane 2). Duplicate gels were transferred onto NC membranes and incubated with different Tv (+) (Lanes 3–12) and Tv (−) (Lanes 13–16) patient sera at 1:25 dilution. Arrowhead points to the recombinant protein band TvLEGU-2r (∼85 kDa). (<b>B</b>) Ponceau red (PR)-stained NC membranes showing the patterns of proteins present in 11 VWs from Tv (+) (Lanes 1–11) and 11 VWs from Tv (−−) patients (Lanes 12–22), which were precipitated by TCA, separated by SDS-PAGE using 10% polyacrylamide gels, and transferred onto NC membranes. (<b>C</b>) WB assays using the anti-TvLEGU-2r (1:1000 dilution) antibody to incubate the NC membranes described in (<b>B</b>). Arrowheads show the positions of the native TvLEGU-2 protein bands (∼49 and ∼29 kDa).</p>
Full article ">
16 pages, 3194 KiB  
Article
The Cysteine Protease Legumain Is Upregulated by Vitamin D and Is a Regulator of Vitamin D Metabolism in Mice
by Karl Martin Forbord, Meshail Okla, Ngoc Nguyen Lunde, Tatjana Bosnjak-Olsen, Guro Arnekleiv, Daniel Hesselson, Harald Thidemann Johansen, Jonathan C. Y. Tang, Moustapha Kassem, Rigmor Solberg and Abbas Jafari
Cells 2024, 13(1), 36; https://doi.org/10.3390/cells13010036 - 22 Dec 2023
Cited by 1 | Viewed by 1734
Abstract
Legumain is a lysosomal cysteine protease that has been implicated in an increasing amount of physiological and pathophysiological processes. However, the upstream mechanisms regulating the expression and function of legumain are not well understood. Here, we provide in vitro and in vivo data [...] Read more.
Legumain is a lysosomal cysteine protease that has been implicated in an increasing amount of physiological and pathophysiological processes. However, the upstream mechanisms regulating the expression and function of legumain are not well understood. Here, we provide in vitro and in vivo data showing that vitamin D3 (VD3) enhances legumain expression and function. In turn, legumain alters VD3 bioavailability, possibly through proteolytic cleavage of vitamin D binding protein (VDBP). Active VD3 (1,25(OH)2D3) increased legumain expression, activity, and secretion in osteogenic cultures of human bone marrow stromal cells. Upregulation of legumain was also observed in vivo, evidenced by increased legumain mRNA in the liver and spleen, as well as increased legumain activity in kidneys from wild-type mice treated with 25(OH)D3 (50 µg/kg, subcutaneously) for 8 days compared to a control. In addition, the serum level of legumain was also increased. We further showed that active legumain cleaved purified VDBP (55 kDa) in vitro, forming a 45 kDa fragment. In vivo, no VDBP cleavage was found in kidneys or liver from legumain-deficient mice (Lgmn−/−), whereas VDBP was cleaved in wild-type control mice (Lgmn+/+). Finally, legumain deficiency resulted in increased plasma levels of 25(OH)D3 and total VD3 and altered expression of key renal enzymes involved in VD3 metabolism (CYP24A1 and CYP27B1). In conclusion, a regulatory interplay between VD3 and legumain is suggested. Full article
(This article belongs to the Topic Osteoimmunology and Bone Biology)
Show Figures

Figure 1

Figure 1
<p><b>Vitamin D3 increases legumain expression, activity, and secretion in pre-osteoblastic cells.</b> (<b>A</b>) The nucleotide sequence of the LGMN gene promoter region with annotations of potential vitamin D-responsive elements (VDRE; red) relative to the transcription start site (TSS). (<b>B</b>–<b>F</b>) Human BMSC-TERT cells (20,000 cells/cm<sup>2</sup>) were incubated with 1,25(OH)<sub>2</sub>D<sub>3</sub> (<b>B</b>–<b>F</b>; 10, 50 or 100 nM), 25(OH)D<sub>3</sub> (<b>C</b>–<b>F</b>; 100, 250, 500 or 1000 nM) or an equal volume of ethanol (control, 0 nM) in osteoblast induction medium for seven days before harvesting. (<b>B</b>) Legumain mRNA expression relative to housekeeping control (GAPDH) (2<sup>−ΔΔCT</sup>; n = 3). (<b>C</b>) One representative immunoblot of legumain (proform 56 kDa, mature form 36 kDa) and GAPDH (housekeeping) in cell lysates (n = 3). (<b>D</b>) Quantification of the 36 kDa mature legumain immunoband (IB) intensity as arbitrary units (ARBU) relative to GAPDH in immunoblots represented in C (n = 3). (<b>E</b>) Legumain activity (dF/s) in cell lysates adjusted for the total protein concentration (µg/mL) (n = 6–9). (<b>F</b>) Secreted legumain (pg/mL) in conditioned media measured by ELISA and adjusted for the total protein concentration in the corresponding cell lysates (n = 3–5). (<b>B</b>,<b>D</b>–<b>F</b>) Data represent mean ± SEM. (<b>B</b>,<b>D</b>) Kruskal–Wallis test. (<b>E</b>,<b>F</b>) One-way ANOVA. * <span class="html-italic">p</span> &lt; 0.05 vs. 0 nM 1,25(OH)<sub>2</sub>D<sub>3</sub> or 25(OH)D<sub>3</sub>. Numbers (n) represent individual biological replicates.</p>
Full article ">Figure 2
<p><b>Treatment with 25(OH)D<sub>3</sub> increases legumain levels and activity in wild-type mice.</b> Wild-type mice (<span class="html-italic">Lgmn</span><sup>+/+</sup>) were treated with 50 µg/kg 25(OH)D<sub>3</sub> (n = 7) or an equal volume vehicle (n = 7, control) subcutaneously every two to three days (four times in total). Tissues were harvested 24 h after the final injection (day 8). (<b>A</b>) Legumain mRNA expression relative to the geometric mean of CT values of four housekeeping controls in kidney, liver, and spleen (2<sup>−ΔΔCT</sup>; n = 5). (<b>B</b>) One representative immunoblot of legumain and GAPDH in kidney, liver, and spleen (n = 3). (<b>C</b>) Quantification of the 36 kDa mature legumain immunoband (IB) intensity as arbitrary units (ARBU) relative to GAPDH (housekeeping) in kidney, liver, and spleen from immunoblots represented in (<b>C</b>) (n = 3). (<b>D</b>) Legumain activity (dF/s) in kidney, liver, and spleen adjusted for total protein concentration (μg/mL, n = 5). (<b>E</b>) Legumain plasma concentration (ng/mL) measured by ELISA (n = 5). (<b>F</b>) Correlation between legumain (ng/mL and 1,25(OH)<sub>2</sub>D<sub>3</sub> (pmol/L) concentrations in plasma (n = 5). (<b>A</b>,<b>C</b>,<b>E</b>) Two-tailed unpaired Student’s <span class="html-italic">t</span>-test. (<b>D</b>) Mann–Whitney test. Data represent mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05. (<b>F</b>) Simple linear regression. Numbers (n) represent individual biological replicates.</p>
Full article ">Figure 3
<p><b>Legumain is required for VDBP processing and regulation</b>. (<b>A</b>) Purified VDBP from human plasma (1.9 μM) was incubated in legumain assay buffer (pH 5.8) at 37 °C with or without purified active bovine legumain (2 μM) for 5 h before gel electrophoresis and immunoblotting of VDBP (n = 1). (<b>B</b>–<b>H</b>) Wild-type (<span class="html-italic">Lgmn</span><sup>+/+</sup>) and legumain-deficient (<span class="html-italic">Lgmn</span><sup>−/−</sup>) mice were treated with 50 µg/kg 25(OH)D<sub>3</sub> (n = 6–7) or an equal volume vehicle (n = 7, control) subcutaneously every two to three days (four times in total). Tissues were harvested 24 h after the final injection (day 8). (<b>B</b>) One representative immunoblot of VDBP and GAPDH (housekeeping) in kidney and liver (n = 4). (<b>C</b>–<b>F</b>) Quantification of VDBP immunoband (IB) intensity as arbitrary units (ARBU) relative to GAPDH in immunoblots represented in (<b>B</b>) (n = 4). (<b>C</b>) Hepatic VDBP 45 kDa immunoband. (<b>D</b>) Renal VDBP 45 kDa immunoband. (<b>E</b>) Hepatic VDBP 55 kDa immunoband. (<b>F</b>) Renal VDBP 55 kDa immunoband. (<b>G</b>) Plasma VDBP concentration (μg/mL) was measured by ELISA (n = 6–7). (<b>H</b>) Hepatic VDBP mRNA expression relative to the geometric mean of CT values of four housekeeping controls (2<sup>−ΔΔCT</sup>, n = 5). (<b>C</b>–<b>H</b>) Data represent mean ± SEM. Two-way ANOVA. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. different genotype, same treatment. Numbers (n) represent individual biological replicates.</p>
Full article ">Figure 4
<p><b>Legumain deficiency alters plasma levels of vitamin D metabolites and induces changes in renal expression of vitamin D-metabolizing enzymes.</b> Wild-type (<span class="html-italic">Lgmn</span><sup>+/+</sup>) and legumain-deficient (<span class="html-italic">Lgmn</span><sup>−/−</sup>) mice were treated with 50 µg/kg 25(OH)D<sub>3</sub> (n = 6–7) or an equal volume vehicle (n = 7, control) subcutaneously every two to three days (four times in total). Tissues were harvested 24 h after the final injection (day 8). (<b>A</b>–<b>D</b>) Vitamin D<sub>3</sub> metabolites in plasma were analyzed by LC-MS/MS. (<b>A</b>) Total plasma concentration of vitamin D<sub>3</sub> metabolites (25(OH)D<sub>3</sub>, 1,25(OH)<sub>2</sub>D<sub>3</sub>, and 24,25(OH)<sub>2</sub>D<sub>3</sub>) (nmol/L, n = 4–5). (<b>B</b>) Plasma 25(OH)D<sub>3</sub> concentration (nmol/L, n = 5). (<b>C</b>) Plasma 1,25(OH)<sub>2</sub>D<sub>3</sub> concentration (pmol/L, n = 4–5). (<b>D</b>) Plasma 24,25(OH)<sub>2</sub>D<sub>3</sub> concentration (nmol/L, n = 5). (<b>E</b>,<b>F</b>) Renal CYP24A1 (<b>E</b>) and CYP27B1 (<b>F</b>) mRNA expressions relative to the geometric mean of CT values of four housekeeping controls (2<sup>−ΔΔCT</sup>; n = 5). Data represent mean ± SEM. (<b>A</b>–<b>D</b>). Two-way ANOVA. (<b>E</b>,<b>F</b>) Two-way ANOVA on ΔCT values. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. same genotype, different treatment. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. different genotype, same treatment. Numbers (n) represent individual biological replicates.</p>
Full article ">Figure 5
<p><b>Graphical representation of the suggested interplay between vitamin D and legumain. Left panel</b>: Vitamin D (VD<sub>3</sub>) promotes legumain expression and activity through transcriptional upregulation of the legumain gene (LGMN). The free fraction of circulating VD<sub>3</sub> metabolites diffuse through plasma membranes. 25-hydroxyvitamin D (25(OH)D<sub>3</sub>) is hydroxylated by 1α-hydroxylase (CYP27B1), forming the active metabolite 1α,25-dihydroxyvitamin D (1,25(OH)<sub>2</sub>D<sub>3</sub>). 1,25(OH)<sub>2</sub>D<sub>3</sub> binds to the nuclear vitamin D receptor (VDR) and promotes transcription of legumain (LGMN). Synthesized prolegumain is either sorted and activated in the endolysosomal system or released to the extracellular environment. <b>Right panel</b>: In the proximal tubular epithelium, 25(OH)D<sub>3</sub> bound to vitamin D binding protein (VDBP) is internalized from the tubular lumen through a megalin/cubilin-mediated process. The vitamin D metabolite is released, enabling subsequent hydroxylation by 1α-hydroxylase (CYP27B1) or 24-hydroxylase (CYP24A1), and VDBP is cleaved by legumain in the endolysosomal system. VDBP cleavage by legumain might be important in controlling the systemic level of vitamin D metabolites. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 11 December 2023).</p>
Full article ">
24 pages, 14241 KiB  
Article
Pigmented Microbial Extract (PMB) from Exiguobacterium Species MB2 Strain (PMB1) and Bacillus subtilis Strain MB1 (PMB2) Inhibited Breast Cancer Cells Growth In Vivo and In Vitro
by Deepa R. Bandi, Ch M. Kumari Chitturi, Jamuna Bai Aswathanarayan, Prashant Kumar M. Veeresh, Venugopal R. Bovilla, Olga A. Sukocheva, Potireddy Suvarnalatha Devi, Suma M. Natraj and SubbaRao V. Madhunapantula
Int. J. Mol. Sci. 2023, 24(24), 17412; https://doi.org/10.3390/ijms242417412 - 12 Dec 2023
Viewed by 1306
Abstract
Breast cancer (BC) continues to be one of the major causes of cancer deaths in women. Progress has been made in targeting hormone and growth factor receptor-positive BCs with clinical efficacy and success. However, little progress has been made to develop a clinically [...] Read more.
Breast cancer (BC) continues to be one of the major causes of cancer deaths in women. Progress has been made in targeting hormone and growth factor receptor-positive BCs with clinical efficacy and success. However, little progress has been made to develop a clinically viable treatment for the triple-negative BC cases (TNBCs). The current study aims to identify potent agents that can target TNBCs. Extracts from microbial sources have been reported to contain pharmacological agents that can selectively inhibit cancer cell growth. We have screened and identified pigmented microbial extracts (PMBs) that can inhibit BC cell proliferation by targeting legumain (LGMN). LGMN is an oncogenic protein expressed not only in malignant cells but also in tumor microenvironment cells, including tumor-associated macrophages. An LGMN inhibition assay was performed, and microbial extracts were evaluated for in vitro anticancer activity in BC cell lines, angiogenesis assay with chick chorioallantoic membrane (CAM), and tumor xenograft models in Swiss albino mice. We have identified that PMB from the Exiguobacterium (PMB1), inhibits BC growth more potently than PMB2, from the Bacillus subtilis strain. The analysis of PMB1 by GC-MS showed the presence of a variety of fatty acids and fatty-acid derivatives, small molecule phenolics, and aldehydes. PMB1 inhibited the activity of oncogenic legumain in BC cells and induced cell cycle arrest and apoptosis. PMB1 reduced the angiogenesis and inhibited BC cell migration. In mice, intraperitoneal administration of PMB1 retarded the growth of xenografted Ehrlich ascites mammary tumors and mitigated the proliferation of tumor cells in the peritoneal cavity in vivo. In summary, our findings demonstrate the high antitumor potential of PMB1. Full article
(This article belongs to the Special Issue Hormone Signaling in Cancers and Cancer-Promoting Pathologies)
Show Figures

Figure 1

Figure 1
<p>Microbial extracts PMB1 and PMB2 inhibited LGMN activity ex vivo. Inhibition of LGMN activity by microbial extracts PMB1 and PMB2 was performed as detailed in the Methods (<a href="#sec4-ijms-24-17412" class="html-sec">Section 4</a>). A dose-dependent increase in the percentage inhibition of LGMN activity was observed with PMB1 and PMB2 extracts. IA (positive control) inhibited LGMN activity by &gt;90% at 750 µM and 1500 µM concentrations (Insert). Statistical analysis was performed using the DMC Test. ****—indicates <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 2
<p>Microbial extracts PMB1 and PMB2 inhibited BC cell growth without affecting the viability of normal lung epithelial cells BEAS-2B. PMB1 more effectively reduced the viability of MDA-MB-468 (<b>A</b>), MDA-MB-231 (<b>D</b>), and MCF-7 (<b>G</b>) BC cells compared to PMB2 effects (<b>B</b>,<b>E</b>,<b>H</b>). Photomicrograph analysis at 10× showed dose-dependent changes in the morphology of BC cells treated with PMB1 (<b>C</b>,<b>F</b>,<b>I</b>) &amp; treatment of normal human lung epithelial cell line BEAS-2B with PMB1 increased cell number more evidently compared to PMB2 (<b>J</b>–<b>L</b>). Dimethyl sulfoxide (DMSO; 1%) was used as the vehicle control. Statistical analysis was performed using the DMC test. *, **, ***, **** indicates <span class="html-italic">p</span> &lt; 0.05, &lt;0.01, 0.001, 0.0001, respectively; “ns” indicates “non significant”.</p>
Full article ">Figure 3
<p>Microbial extract PMB1 inhibited LGMN activity and reduced the migration of TNBC cell line MDA-MB-468. (<b>A</b>) Expression of LGMN in MDA-MB-468 cells treated with microbial extract PMB1 for 48 h showed a significant reduction at 250 µg/mL and 1000 µg/mL concentration. (<b>B</b>) The inhibition in LGMN activity compared to untreated cells was 50.6, 72.6 and 72.4% in 100 µg/mL, 250 µg/mL, and 1000 µg/mL treated cells, respectively; microbial extract PMB1 mitigated the migration of MDA-MB-468 cells (<b>C</b>–<b>F</b>). Photomicrographs (30×) of untreated MDA-MB-468 cells and the cells exposed to DMSO (1%), positive control DADS (750 µM), and increasing concentration (500 µg/mL, 1000 µg/mL, and 2000 µg/mL) of PMB1 at 24 and 48 h. PMB1 inhibited the migration of MDA-MB-468 cells, as evidenced by a minimal decrease in the scratch compared to untreated control and vehicle-treated cells. *, ***, indicates <span class="html-italic">p</span> &lt; 0.05, &lt;0.001, respectively; “ns” indicates “non significant”.</p>
Full article ">Figure 4
<p>Microbial extract PMB1 inhibited angiogenesis in a dose-dependent manner in chick chorioallantoic membrane (CAM) assay (<b>A</b>–<b>C</b>) Photograph of CAM exposed to control and vehicle PBS at 0, 24 and 48 h (<b>A</b>), 500 µg/mL of microbial extract PMB1 (<b>B</b>) and 1000 µg/mL microbial extract PMB1 (<b>C</b>). A dose-dependent decrease in the number as well as size of the blood vessels was observed with PMB1 treatment (<b>C</b>).</p>
Full article ">Figure 5
<p>PMB1 induced apoptosis in the MDA-MB-468 cell line. Exposure of MDA-MB-468 cells to vehicle control (VC; 1% DMSO) did not induce any significant apoptosis (<b>A</b>); however, treatment with positive control 750 µM DADS moderately elevated cells undergoing apoptotic death. Microbial extract PMB1-treated cells (500 µg/mL, 1000 µg/mL, and 2000 µg/mL) showed a dose-dependent increase in the percentage of apoptotic cells (<b>B</b>). Photomicrographs (10×) of cells treated with microbial extract PMB1 (500 µg/mL, 1000 µg/mL and 2000 µg/mL). A dose-dependent increase in the apoptotic cells was observed; Apoptosis detection in microbial extract PMB1-treated (500 µg/mL, 1000 µg/mL, and 2000 µg/mL) MDA-MB-468 cells using ethidium bromide and acridine orange staining. The images obtained using 2 different channels were merged to obtain a combined image, which emitted green and orange cells. The live cells take up acridine orange which stains the cells green, while the apoptotic cells, whose membrane integrity is lost and nucleus is exposed take up the ethidium bromide and appear orange when photomicrographed under fluorescence microscope. (<b>C</b>). Morphological changes in the MDA-MB-468 cells upon treatment with microbial extract PMB1 (100 µg/mL, 250 µg/mL, and 1000 µg/mL) for 48 h showed a major difference in the cell size and morphology between control and PMB1-treated samples (100 µg/mL, 250 µg/mL, and 1000 µg/mL) (<b>D</b>). Comparison of change in cell number of MDA-MB-468 cells treated with microbial extract (100 µg/mL, 250 µg/mL, and 1000 µg/mL) for 48 h and control using the DMC test (<b>E</b>). Comparison of the viability of MDA-MB-468 cells treated with microbial extract (100 µg/mL, 250 µg/mL, and 1000 µg/mL) for 48 h with controls using the DMC test (<b>F</b>). *, **, indicates <span class="html-italic">p</span> &lt; 0.05, &lt;0.01, respectively; “ns” indicates “non significant”.</p>
Full article ">Figure 6
<p>Microbial extract PMB1 exhibited anticancer activity in EAC xenografts in mice in vivo. (<b>A</b>) Body weight changes. (<b>B</b>) Comparison of the tumor volume on the day of sacrifice (Day 20) between different treatment groups and controls. (<b>C</b>) Viability of EAC cells collected from peritoneal fluid. (<b>D</b>) A comparison of the number of EAC cells collected from peritoneal fluid of different treatment groups showed a moderate increase in the number of apoptotic cells. (<b>E</b>) Comparison of % of apoptotic cells in different treatment groups in the peritoneal fluid of Swiss albino mice treated with PMB1 compared to control untreated EAC mice. (<b>F</b>) Apoptosis detection in EAC cells collected from mice peritoneum on the day of sacrifice in control, positive control, and microbial extract PMB1-treated group. Green color indicates live cells whereas the Red color indicates the apoptotic cells (<b>G</b>) The Kaplan-Meier survival analysis for the calculation of median survival time showed beneficial outcomes with PMB1-treated mice compared to control animals. (<b>H</b>) H&amp;E staining of liver and kidney tissues (20×) in control, cisplatin, and microbial extract PMB1-treated groups. The tumor volume was measured using vernier calipers on every alternative day. (<b>I</b>,<b>J</b>) Tumors excised from the mice on the 25th day of the treatment. *, **, **** indicates <span class="html-italic">p</span> &lt; 0.05, &lt;0.01, 0.0001, respectively.</p>
Full article ">
14 pages, 2232 KiB  
Review
Lysosomal-Cleavable Peptide Linkers in Antibody–Drug Conjugates
by Seetharamsing Balamkundu and Chuan-Fa Liu
Biomedicines 2023, 11(11), 3080; https://doi.org/10.3390/biomedicines11113080 - 16 Nov 2023
Cited by 9 | Viewed by 5647
Abstract
Antibody–drug Conjugates (ADCs) are a powerful therapeutic modality for cancer treatment. ADCs are multi-functional biologics in which a disease-targeting antibody is conjugated to an effector payload molecule via a linker. The success of currently used ADCs has been largely attributed to the development [...] Read more.
Antibody–drug Conjugates (ADCs) are a powerful therapeutic modality for cancer treatment. ADCs are multi-functional biologics in which a disease-targeting antibody is conjugated to an effector payload molecule via a linker. The success of currently used ADCs has been largely attributed to the development of linker systems, which allow for the targeted release of cytocidal payload drugs inside cancer cells. Many lysosomal proteases are over expressed in human cancers. They can effectively cleave a variety of peptide sequences, which can be exploited for the design of ADC linker systems. As a well-established linker, valine-citrulline-p-aminobenzyl carbamate (ValCitPABC) is used in many ADCs that are already approved or under preclinical and clinical development. Although ValCitPABC and related linkers are readily cleaved by cathepsins in the lysosome while remaining reasonably stable in human plasma, many studies have shown that they are susceptible to carboxylesterase 1C (Ces1C) in mouse and rat plasma, which hinders the preclinical evaluation of ADCs. Furthermore, neutropenia and thrombocytopenia, two of the most commonly observed dose-limiting adverse effects of ADCs, are believed to result from the premature hydrolysis of ValCitPABC by human neutrophil elastase. In addition to ValCitPABC, the GGFG tetrapeptidyl-aminomethoxy linker is also cathepsin-cleavable and is used in the highly successful ADC drug, DS8201a. In addition to cathepsin-cleavable linkers, there is also growing interest in legumain-sensitive linkers for ADC development. Increasing plasma stability while maintaining lysosomal cleavability of ADC linkers is an objective of intensive current research. This review reports recent advances in the design and structure–activity relationship studies of various peptide/peptidomimetic linkers in this field. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) Fate of an ADC before and after internalization. Premature cleavage of the linker in extracellular matrix is often associated with off-target toxicity. Antigen-mediated endocytosis delivers ADC in the endosomal–lysosomal system and lysosomal linker cleavage releases the drug, which acts to exert its cytotoxicity. (<b>B</b>) Effect of amino acid composition in the linker peptide and substitution on the benzene ring of PABC on linker stability in mouse plasma.</p>
Full article ">Figure 2
<p>Various Linker payload designs for faster lysosomal cleavage and improved plasma stability. (<b>A</b>) Peptidomimetic linkers specifically cleaved by cathepsin B; (<b>B</b>) modifications to central PABC ring.</p>
Full article ">Figure 3
<p>Various Linker payload designs for faster lysosomal cleavage and improved plasma stability. (<b>A</b>) Tandem cleavable linkers’ glucuronide group masks the linker system to maintain stability in extracellular environment; (<b>B</b>) Aryalsulfatase A (ARSA) and β-galactosidase dual cleavable 3-<span class="html-italic">O</span>-sulfo-β-galactose linker.</p>
Full article ">Figure 4
<p>Structure and linker cleavage mechanism of trastuzumab deruxtecan (Enhertu).</p>
Full article ">Figure 5
<p>Asn-containing peptide linkers with improved selectivity towards legumain.</p>
Full article ">
28 pages, 2830 KiB  
Review
Extracellular Cysteine Proteases of Key Intestinal Protozoan Pathogens—Factors Linked to Virulence and Pathogenicity
by Raúl Argüello-García, Julio César Carrero and M. Guadalupe Ortega-Pierres
Int. J. Mol. Sci. 2023, 24(16), 12850; https://doi.org/10.3390/ijms241612850 - 16 Aug 2023
Cited by 3 | Viewed by 1984
Abstract
Intestinal diseases caused by protistan parasites of the genera Giardia (giardiasis), Entamoeba (amoebiasis), Cryptosporidium (cryptosporidiosis) and Blastocystis (blastocystosis) represent a major burden in human and animal populations worldwide due to the severity of diarrhea and/or inflammation in susceptible hosts. These pathogens interact with [...] Read more.
Intestinal diseases caused by protistan parasites of the genera Giardia (giardiasis), Entamoeba (amoebiasis), Cryptosporidium (cryptosporidiosis) and Blastocystis (blastocystosis) represent a major burden in human and animal populations worldwide due to the severity of diarrhea and/or inflammation in susceptible hosts. These pathogens interact with epithelial cells, promoting increased paracellular permeability and enterocyte cell death (mainly apoptosis), which precede physiological and immunological disorders. Some cell-surface-anchored and molecules secreted from these parasites function as virulence markers, of which peptide hydrolases, particularly cysteine proteases (CPs), are abundant and have versatile lytic activities. Upon secretion, CPs can affect host tissues and immune responses beyond the site of parasite colonization, thereby increasing the pathogens’ virulence. The four intestinal protists considered here are known to secrete predominantly clan A (C1- and C2-type) CPs, some of which have been characterized. CPs of Giardia duodenalis (e.g., Giardipain-1) and Entamoeba histolytica (EhCPs 1-6 and EhCP112) degrade mucin and villin, cause damage to intercellular junction proteins, induce apoptosis in epithelial cells and degrade immunoglobulins, cytokines and defensins. In Cryptosporidium, five Cryptopains are encoded in its genome, but only Cryptopains 4 and 5 are likely secreted. In Blastocystis sp., a legumain-activated CP, called Blastopain-1, and legumain itself have been detected in the extracellular medium, and the former has similar adverse effects on epithelial integrity and enterocyte survival. Due to their different functions, these enzymes could represent novel drug targets. Indeed, some promising results with CP inhibitors, such as vinyl sulfones (K11777 and WRR605), the garlic derivative, allicin, and purified amoebic CPs have been obtained in experimental models, suggesting that these enzymes might be useful drug targets. Full article
(This article belongs to the Special Issue Microbial Proteases: Structure, Function and Role in Pathogenesis)
Show Figures

Figure 1

Figure 1
<p>CPs from <span class="html-italic">Giardia duodenalis</span> trophozoites interact with multiple targets in the small intestine. Experimental models of trophozoites–epithelial cell interactions indicate that the parasite releases CPs, mainly cathepsin B-type, including Giardipain-1 (GL50803_14019), GL16160 and GL16779 and non-canonical CPs (e.g., VSP9B10A). This latter CP may serve, when expressed, to divert the immune system (1) or may cause damage to the epithelium due to a loss of cell–cell junction and cytotoxicity in the cell (2). Secreted CPs have been shown to degrade substrates, such as mucin 2, an important component of intestinal mucus (3), enabling trophozoite adhesion to microvilli. Direct damage to epithelial cell integrity by CPs may include disruption of the cytoskeletal microvillus-resident protein villin (4) and the disruption of tight junction proteins, such as ZO-1 and claudins, also involving adherens junction proteins including β-catenin or E-cadherin (5). Soluble elements of the innate immune response, including secretory IgA (6), produced by plasmatic B-cells, defensins (7) and IL-8 (8), both produced by epithelial cells, of which the latter works as neutrophil attractant, might be degraded by giardial CPs. These enzymes also provoke alterations in the microbiome of the small intestine, leading to dysbiosis (9), while bacterial translocation from the luminal to the intraepithelial compartment (10) may be promoted via the degradation of intercellular junctions by CPs as mentioned. Recent studies suggest that giardial CPs might be secreted after removal of N-terminal prodomain and inclusion into membrane-bound extracellular vesicles (EVs), mainly exosomes (green-filled circles).</p>
Full article ">Figure 2
<p><span class="html-italic">Entamoeba histolytica</span> CPs (EhCPs) are central to invasion, pathogenicity and immune evasion during intestinal and extraintestinal amoebiasis. Once the amoebic trophozoite reaches the mucus layer in the large intestine (<b>a</b>), EhCPs released in extracellular vesicles (EVs) can degrade the mucin backbone and secretory IgA antibodies (sIgA), which facilitates initiation of mucosal invasion (degradation is indicated by red rays). Intriguingly, the participation of EhCPs in the secretagogue capacity of the amoeba has also been described (activation is indicated by the blue rays). During trophozoite contact with the apical region of the intestinal epithelium (<b>b</b>), EhCPs degrade components of the extracellular matrix and villin in the apical region of enterocytes, eroding the epithelium and activating signaling pathways that lead to nuclear translocation of the transcription factor NF-κB, the inflammasome assembly and the expression of proinflammatory cytokines. The penetration of the amoeba through the epithelium (<b>c</b>) occurs in the intercellular spaces by EhCPs degradation of tight junctions, adhesion junctions and desmosomes components. At the same time, the parasite induces the death of enterocytes by trogocytosis and apoptosis, with EhCPs participating in the former. Since the amoeba is in the submucosa (<b>d</b>), the EhCPs can activate mast cells to produce IL-8, while they can degrade cytokines such as Pro-IL-18, or in contrast, activate them as in the case of Pro-IL-1B. At this point, EhCPs can also destroy intestinal nervous tissue (neurons/axons), affecting its physiology. During the invasion of the tissue, the amoeba also comes in contact with blood components (<b>e</b>), such as complement and IgG antibodies, which are degraded by EhCPs. At this point, in very sporadic cases and for reasons not yet understood, amoebae can migrate through the portal vein to the liver (<b>f</b>), where EhCPsbreak down hemoglobin to use iron. Finally, once in the liver (<b>g</b>), EhCPs contribute to tissue damage through the degradation of cell matrix components, enterocytes, and recruited immune cells, leading to the development of amoebic liver abscesses.</p>
Full article ">Figure 3
<p>Protein structure models representing the Cryptopain family. The protein models of Cryptopain-1 (<b>A</b>), -2 (<b>B</b>), -3 (<b>C</b>), -4 (<b>D</b>) and -5 (<b>E</b>) were obtained using the I-Tasser server (<a href="https://zhanggroup.org/I-TASSER/" target="_blank">https://zhanggroup.org/I-TASSER/</a>; accessed dates: 28 February 2023, 3 March 2023, 22 and 28 July 2023), and the domains were identified with the InterPro platform (<a href="https://www.ebi.ac.uk/interpro/" target="_blank">https://www.ebi.ac.uk/interpro/</a>; accessed dates: 28 February 2023, 3 March 2023, 22 and 28 July 2023) and are indicated by colors at lower right. The catalytic triad Cys-His-Asn is displayed in ball-and-stick conformation and is magnified within dotted squares. Cryptopains 1-3 are cathepsin L-type and Cryptopains 4 and 5 are cathepsin B-type. From these analyses, Cryptopains 1 and 3 are predicted to be membrane-anchored (possess transmembrane domain), Cryptopain-2 is cytoplasmic (signal peptide and transmembrane domains absent) and Cryptopains 4 and 5 are secreted (possess signal peptide). Protein models were visualized and edited using the UCSF Chimera server v1.10.17.</p>
Full article ">Figure 4
<p>Blastopain-1 and other cysteine proteases from <span class="html-italic">Blastocystis</span>. Possible role in pathogenesis and immune evasion. (<b>A</b>) Protein structure models of zymogen and active forms of a secreted, Cathepsin B (Blastopain-1) and legumain from <span class="html-italic">Blastocystis</span> sp. The catalytic triad Cys(red)-His(orange)-Asn (purple) is displayed in <span class="html-italic">ball-and-stick</span> conformation and is magnified within the dotted squares Protein domains were obtained from the InterPro platform and are colored as follows: signal peptides (SP) in magenta; papain-like signature (CatB) in green; cathepsin C prodomain (CatB) and auto-inhibitory C-terminal prodomain (legumain) in blue; hemoglobinase C13 signature (legumain) in cyan; and occluding loop (CatB) in brown. Models were obtained using the I-Tasser server from sequences with a.n. CBK25506-2 (CatB) and CBK21815-2 (legumain). (<b>B</b>) Proposed roles of cysteine proteases from <span class="html-italic">Blastocystis</span>. The cyst (C) form precedes the vegetative vacuolar (V) form that alternates with other entities (ameboid, granular) at intestinal lumen where cysteine proteases (CPs) may be secreted and causes effects at different levels: (1) parasites attached at intercellular junctions may release CPs that degrade junctional proteins such as ZO1- and claudins, promoting increased epithelial permeability; (2) CPs at intraepithelial compartment might induce upregulation of proinflammatory cytokines in Monocytes/Macrophages (Mϕ) and T lymphocytes; (3) epithelial cells exposed to CPs produce IL-8, a potent chemoattractant for Neutrophils and Polymorphonuclear cells; and (4) disruption of intercellular junctions along to a likely activation of Caspase-3 pathway may result in programmed cell death (apoptosis). Also, secreted CPs are able to degrade secretory IgA in vitro (5), an important effector in the mucosal system that has been observed at increased levels in symptomatic cases of Blastocystosis.</p>
Full article ">Figure 5
<p>Mechanisms of inhibition of CPs from intestinal protozoa by synthetic and natural compounds. Protein models of secreted CPs were obtained by homology modeling using the Phyre2 server. (<b>A</b>) Blockade of the active site of EhCP1 by the vinyl sulfone K11777. Prediction model from SwissDock server (<a href="http://www.swissdock.ch/docking" target="_blank">http://www.swissdock.ch/docking</a>; accessed dates: 28 March 2023 and 11 July 2023) of a favored docking position (ΔG = −7.2436kCal/mol) of K11777 (in cyan, displayed in <span class="html-italic">stick</span> conformation) at the vicinity of the catalytic triad of EhCP1 displayed in ball-and-stick conformation (cysteine in red, histidine in orange and asparagine in purple). K11777 structure was obtained in SMILES format with further energy minimization using Avogadro suite v1.2. As reference, chemical structures of vinyl sulfone inhibitors K11777 and WRR483 are shown on the right. (<b>B</b>) Modification of active site cysteine from Giardipain-1 by allicin. The protein backbone shows the positions of catalytic residues that are displayed as described above. Upon interaction with allicin (ALC), the catalytic cysteine is converted into S-allylthiocysteine (SATC), which lacks nucleophilic nature as the cysteine thiol, thereby inactivating Giardipain-1. In this case, other thiol-disulfide exchange reactions could proceed with allosteric cysteines, perturbing enzyme activity. Protein models were visualized and edited using the UCSF Chimera server v1.10.17.</p>
Full article ">
17 pages, 3791 KiB  
Article
Pharmacological Inhibition of the Asparaginyl Endopeptidase (AEP) in an Alzheimer’s Disease Model Improves the Survival and Efficacy of Transplanted Neural Stem Cells
by Qing Cheng, Xiaoli Ma, Jingjing Liu, Xuemei Feng, Yan Liu, Yanxia Wang, Wenwen Ni and Mingke Song
Int. J. Mol. Sci. 2023, 24(9), 7739; https://doi.org/10.3390/ijms24097739 - 23 Apr 2023
Cited by 3 | Viewed by 1844
Abstract
Stem-cell-based therapy is very promising for Alzheimer’s disease (AD), yet has not become a reality. A critical challenge is the transplantation microenvironment, which impacts the therapeutic effect of stem cells. In AD brains, amyloid-beta (Aβ) peptides and inflammatory cytokines continuously poison the tissue [...] Read more.
Stem-cell-based therapy is very promising for Alzheimer’s disease (AD), yet has not become a reality. A critical challenge is the transplantation microenvironment, which impacts the therapeutic effect of stem cells. In AD brains, amyloid-beta (Aβ) peptides and inflammatory cytokines continuously poison the tissue microenvironment, leading to low survival of grafted cells and restricted efficacy. It is necessary to create a growth-supporting microenvironment for transplanted cells. Recent advances in AD studies suggest that the asparaginyl endopeptidase (AEP) is a potential intervention target for modifying pathological changes. We here chose APP/PS1 mice as an AD model and employed pharmacological inhibition of the AEP for one month to improve the brain microenvironment. Thereafter, we transplanted neural stem cells (NSCs) into the hippocampus and maintained therapy for one more month. We found that inhibition of AEPs resulted in a significant decrease of Aβ, TNF-α, IL-6 and IL-1β in their brains. In AD mice receiving NSC transplantation alone, the survival of NSCs was at a low level, while in combination with AEP inhibition pre-treatment the survival rate of engrafted cells was doubled. Within the 2-month treatment period, implantation of NSCs plus pre-inhibition of the AEP significantly enhanced neural plasticity of the hippocampus and rescued cognitive impairment. Neither NSC transplantation alone nor AEP inhibition alone achieved significant efficacy. In conclusion, pharmacological inhibition of the AEP ameliorated brain microenvironment of AD mice, and thus improved the survival and therapeutic efficacy of transplanted stem cells. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Figure 1
<p>Mouse neural stem cells (NSCs) transfected with green fluorescent protein (GFP). (<b>a</b>) Bright field images of neurospheres in the suspension culture and dispersed NSCs in the adherent monolayer culture. Scale bar, 200 μm. (<b>b</b>) NSCs and GFP-NSCs stably expressed SOX2, a persistent marker for neural stem cell multipotency. Scale bar, 25 μm. (<b>c</b>) GFP-NSCs and the native NSCs differentiated into neurons (MAP2 positive) and GFAP positive astrocytes. Scale bar, 25 μm. (<b>d</b>) Cell viability of NSCs after a 48 h incubation with TNF-α (10 ng/mL), IL-6 (10 ng/mL), IL-1β (50 ng/mL), Aβ<sub>1–40</sub> (15 μM) and Aβ<sub>1–42</sub> (15 μM), respectively. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = three independent experiments.</p>
Full article ">Figure 2
<p>The enzymatic activity of the asparaginyl endopeptidase (AEP) and proinflammatory cytokines in whole brain tissue of mice. (<b>a</b>) Activity of AEPs detected in brains of WT and APP/PS1 mice, 4 to 8 months old. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = 4–5 mice per group. (<b>b</b>) Expression of TNF-α, IL-6 and IL-1β in brains of 5-to-8-month-old mice. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = 4–5 mice per group.</p>
Full article ">Figure 3
<p>Pharmacological inhibition of the AEP with δ-secretase inhibitor 11. (<b>a</b>) The concentration-response of δ-secretase inhibitor 11 on AEP activity, <span class="html-italic">n</span> = 5 tests. (<b>b</b>) Brain AEP activity of APP/PS1 mice after one month of treatment with vehicle or δ-secretase inhibitor 11. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = 5 mice per group. (<b>c</b>) Aβ<sub>1–40</sub> and Aβ<sub>1–42</sub> in brains of APP/PS1 mice treated with vehicle or δ-secretase inhibitor 11 for one month. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = 4–5 mice per group. (<b>d</b>,<b>e</b>) Transcript and secretion levels of TNF-α, IL-6 and IL-1β in brains of APP/PS1 mice after one-month treatment with δ-secretase inhibitor 11. * <span class="html-italic">p</span> &lt; 0.05. (<b>f</b>,<b>g</b>) TNF-α, IL-6 and IL-1β levels after two-month treatment with δ-secretase inhibitor 11. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = 4–5 mice per group.</p>
Full article ">Figure 4
<p>One month of AEP inhibition treatment and the cognitive function of APP/PS1 mice. (<b>a</b>) Over the 5-day acquisition training, the 3 groups of mice showed similar swimming speed. (<b>b</b>) The escape latency of WT mice compared with APP/PS1 mice treated with either δ-secretase inhibitor 11 or vehicle (<span class="html-italic">p</span> &lt; 0.05, two-way ANOVA analysis, <span class="html-italic">n</span> = 16 mice per group). (<b>c</b>,<b>d</b>) The number of crossing platforms and time spent in the target quadrant (probe test), which was performed on day 6 after the acquisition training. * <span class="html-italic">p</span> &lt; 0.05, WT mice compared with APP/PS1 mice treated with either δ-secretase inhibitor 11 or vehicle, one-way ANOVA followed by a Tukey’s post hoc test, <span class="html-italic">n</span> = 16 mice per group. (<b>e</b>) For cell transplantation experiment, the 8-month-old APP/PS1 mice that had received 1 month of treatment with vehicle or the AEP inhibitor were divided into 4 groups: (1) APP/PS1 + vehicle, (2) APP/PS1 + NSCs, (3) APP/PS1 + inhibitor and (4) APP/PS1 + inhibitor + NSCs. The treatment was planned to be extended for one more month. <span class="html-italic">n</span> = eight mice per group.</p>
Full article ">Figure 5
<p>Survival of NSCs two weeks after implantation into the hippocampus of APP/PS1 mice. (<b>a</b>) GFP expressing NSCs (GFP-NSCs) were tracked in the injection sites of AD mice with or without pre-treatment with the AEP inhibitor. Scale bar, 200 μm. (<b>b</b>) Survival rate of NSCs engrafted in brains of the two group mice. * <span class="html-italic">p</span> &lt; 0.05, unpaired Student’s <span class="html-italic">t</span>-test; <span class="html-italic">n</span> = five mice per group.</p>
Full article ">Figure 6
<p>The cognitive function of APP/PS1 mice and neural plasticity in the hippocampus one month after cell transplantation. (<b>a</b>) Over the 5-day acquisition training, the 5 groups of mice showed similar swimming speed. (<b>b</b>) The escape latency of the five groups. * <span class="html-italic">p</span> &lt; 0.05, APP/PS1 mice receiving NSCs + AEP inhibitor treatment compared with mice treated with either NSC alone or δ-secretase inhibitor 11 alone, two-way ANOVA analysis, <span class="html-italic">n</span> = eight mice per group. (<b>c</b>) The number of crossing platforms (probe test). * <span class="html-italic">p</span> &lt; 0.05, APP/PS1 mice receiving NSCs + AEP inhibitor compared with mice treated with either vehicle or AEP inhibitor. (<b>d</b>) Time spent in the target quadrant (probe test). * <span class="html-italic">p</span> &lt; 0.05, APP/PS1 mice receiving NSCs + AEP inhibitor compared with mice treated with either vehicle or NSCs alone. Probe test data were analyzed by one-way ANOVA and a Tukey’s post hoc test, <span class="html-italic">n</span> = eight mice per group. (<b>e</b>) Long-term potentiation (LTP) of fEPSPs induced in the hippocampus of AD mice. * <span class="html-italic">p</span> &lt; 0.05, the LTP magnitude (from 5 to 60 min) was higher in AD mice receiving NSCs + AEP inhibitor than mice + NSCs alone (two-way ANOVA, <span class="html-italic">n</span> = 6 mice per group). Trace 1 and trace 2 are representative fEPSPs of WT mice recorded at the time point 1 and 2.</p>
Full article ">
15 pages, 2673 KiB  
Article
Legumain Functions as a Transient TrkB Sheddase
by Christoph Holzner, Katharina Böttinger, Constantin Blöchl, Christian G. Huber, Sven O. Dahms, Elfriede Dall and Hans Brandstetter
Int. J. Mol. Sci. 2023, 24(6), 5394; https://doi.org/10.3390/ijms24065394 - 11 Mar 2023
Viewed by 1703
Abstract
While primarily found in endo-lysosomal compartments, the cysteine protease legumain can also translocate to the cell surface if stabilized by the interaction with the RGD-dependent integrin receptor αVβ3. Previously, it has been shown that legumain expression is inversely related to BDNF-TrkB activity. Here [...] Read more.
While primarily found in endo-lysosomal compartments, the cysteine protease legumain can also translocate to the cell surface if stabilized by the interaction with the RGD-dependent integrin receptor αVβ3. Previously, it has been shown that legumain expression is inversely related to BDNF-TrkB activity. Here we show that legumain can conversely act on TrkB-BDNF by processing the C-terminal linker region of the TrkB ectodomain in vitro. Importantly, when in complex with BDNF, TrkB was not cleaved by legumain. Legumain-processed TrkB was still able to bind BDNF, suggesting a potential scavenger function of soluble TrkB towards BDNF. The work thus presents another mechanistic link explaining the reciprocal TrkB signaling and δ-secretase activity of legumain, with relevance for neurodegeneration. Full article
(This article belongs to the Special Issue Lysosomal Proteases and Their Inhibitors)
Show Figures

Figure 1

Figure 1
<p>Preparation of rat TrkB and proBDNF. (<b>A</b>) Domain organization of TrkB. CC1/2: cysteine cluster 1/2; LRM: leucine-rich-motif; Ig1/2: Ig-like domain 1/2; ML: membrane linker; KD: kinase domain. (<b>B</b>) SDS-PAGE of TrkB variants. Lane 1: purified TrkB ECD; Lane 2: purified TrkB Ig2 domain with different glycosylation isoforms. (<b>C</b>) HPLC-MS analysis of the relative fractional abundance of the correct disulfide isoform Cys302-Cys345 in TrkB Ig2. Fractional abundances of the disulfide bonds, as detected by mass spectrometry, are normalized frequencies, i.e., they add up to 100%. (<b>D</b>) SDS-PAGE of (pro-)BDNF. Lane 1: purified proBDNF; Lane 2: furin-activated BDNF; the 70 kDa band corresponds to inactivated furin.</p>
Full article ">Figure 2
<p>Legumain processes TrkB. A+B: Coomassie-stained SDS PAGE gel (<b>A</b>) and corresponding Immunoblot (<b>B</b>) of TrkB ECD and Ig2 domain in the presence (+) and absence (−) of legumain, denoted as LEG. An anti-His6 antibody is used in the Immunoblot to detect the C-terminal His-tag of the uncleaved TrkB variants. M represents the molecular marker lane. (<b>C</b>) Relative fractional abundance of three detected cleavage sites CS1 (Asp385), CS2 (Asn389), and CS3 (Asn391) at pH 5.5 and 7.0. Fractional abundances of the individual cleavage events, as detected by mass spectrometry, are normalized frequencies, i.e., they add up to 100%. (<b>D</b>) Cartoon representation of the TrkB ECD with the three detected cleavage sites CS1, CS2, and CS3 (Asp385, Asn389, and Asn391) in the membrane linker segment with their cleavage predominance indicated by their color intensity.</p>
Full article ">Figure 3
<p>TrkB in complex with BDNF is protected from processing by legumain. M represents the protein marker. In the next three lanes, the presence or absence (+/−) of BDNF and legumain (“LEG”) on the processing of TrkB ECD is tested, followed by three lanes on the processing of TrkB Ig2 domain. The outer right lanes represent controls of BDNF and legumain (LEG) only, migrating at approximately 14 kDa and 36 kDa, respectively. Note the presence of cell medium-derived BSA migrating at 60 kDa. Additionally, in the BDNF sample, inactivated furin is visible at 70 kDa, which was used to activate proBDNF.</p>
Full article ">Figure 4
<p>Legumain-processed TrkB variants bind BDNF. Non-processed TrkB variants bind proBDNF. (<b>A</b>) The legumain-processed TrkB ECD binds BDNF in solution with a K<sub>d</sub> of 188 nM. (<b>B</b>) The legumain-processed TrkB Ig2 binds BDNF in solution with a K<sub>d</sub> of 300 nM. Of note, in both cases, secondary binding events at micromolar concentrations were observed. (<b>C</b>) TrkB ECD binds proBDNF in solution with a K<sub>d</sub> of 306 nM. (<b>D</b>) TrkB Ig2 binds proBDNF in solution with a K<sub>d</sub> of 441 nM. Contrasting the situation with mature BDNF, no secondary binding events were observed.</p>
Full article ">Figure 5
<p>Possible impact of TrkB shedding by Legumain. On the left side, BDNF mediates a heterotetrameric complex with membrane-bound TrkB (TrkB-BDNF)<sub>2</sub>, which prevents shedding by legumain (“LEG”). In the absence of BDNF, TrkB can be shed from the membrane. Soluble TrkB can still bind BDNF, resulting in a soluble (TrkB-BDNF)<sub>2</sub> complex.</p>
Full article ">
22 pages, 1149 KiB  
Review
The Mammalian Cysteine Protease Legumain in Health and Disease
by Rigmor Solberg, Ngoc Nguyen Lunde, Karl Martin Forbord, Meshail Okla, Moustapha Kassem and Abbas Jafari
Int. J. Mol. Sci. 2022, 23(24), 15983; https://doi.org/10.3390/ijms232415983 - 15 Dec 2022
Cited by 20 | Viewed by 3488
Abstract
The cysteine protease legumain (also known as asparaginyl endopeptidase or δ-secretase) is the only known mammalian asparaginyl endopeptidase and is primarily localized to the endolysosomal system, although it is also found extracellularly as a secreted protein. Legumain is involved in the regulation of [...] Read more.
The cysteine protease legumain (also known as asparaginyl endopeptidase or δ-secretase) is the only known mammalian asparaginyl endopeptidase and is primarily localized to the endolysosomal system, although it is also found extracellularly as a secreted protein. Legumain is involved in the regulation of diverse biological processes and tissue homeostasis, and in the pathogenesis of various malignant and nonmalignant diseases. In addition to its proteolytic activity that leads to the degradation or activation of different substrates, legumain has also been shown to have a nonproteolytic ligase function. This review summarizes the current knowledge about legumain functions in health and disease, including kidney homeostasis, hematopoietic homeostasis, bone remodeling, cardiovascular and cerebrovascular diseases, fibrosis, aging and senescence, neurodegenerative diseases and cancer. In addition, this review addresses the effects of some marketed drugs on legumain. Expanding our knowledge on legumain will delineate the importance of this enzyme in regulating physiological processes and disease conditions. Full article
(This article belongs to the Special Issue Peptidases: Role and Function in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Illustration of legumain activation. Inactive prolegumain (56 kDa) is composed of the catalytic domain (light green), activation peptide (AP, grey), and legumain stabilization and activity modulation domain (LSAM, orange), and is stable at a neutral pH. The proenzyme undergoes autocatalytic processing at asparagine 323 (N323) at pH ≤ 5.5 and at aspartate 303/309 (D303/309) at pH ≤ 4.5. Another in trans processing occurs after the KRK289 motif by unknown protease(s). Release of the LSAM domain is mandatory to gain AEP activity (dark green) and the cysteine (C189) in the catalytic site is marked (red dot). In addition, ACP activity is obtained when the LSAM remains electrostatically bound, whereas the AP is removed. A decrease in pH causes protonation and release of the LSAM domain and legumain acquires AEP activity. The amino-terminal cleavage after aspartate 21 (D21) and 25 (D25) is not shown. AEP, aspaparaginyl endopeptidase; ACP, asparaginyl carboxypeptidase. Created by PowerPoint.</p>
Full article ">Figure 2
<p>Illustration of the involvement of legumain in various physiological processes (blue boxes) and diseases (red boxes) in different organs and tissues of the body. See the main text for description and references. Created by BioRender.</p>
Full article ">
14 pages, 2067 KiB  
Article
Identification of Novel Plasma Biomarkers for Abdominal Aortic Aneurysm by Protein Array Analysis
by Jianqiang Wu, Wei Wang, Ting Xie, Zhaoran Chen, Lei Zhou, Xiaohong Song, Haoxuan Kan, Yanze Lv, Lianglin Wu, Fangda Li, Dan Yang, Yuexin Chen, Bao Liu and Yuehong Zheng
Biomolecules 2022, 12(12), 1853; https://doi.org/10.3390/biom12121853 - 12 Dec 2022
Cited by 3 | Viewed by 1886
Abstract
Abdominal aortic aneurysm (AAA) is a potentially life-threatening disease that is common in the aging population. Currently, there are no approved diagnostic biomarkers or therapeutic drugs for AAA. We aimed to identify novel plasma biomarkers or potential therapeutic targets for AAA using a [...] Read more.
Abdominal aortic aneurysm (AAA) is a potentially life-threatening disease that is common in the aging population. Currently, there are no approved diagnostic biomarkers or therapeutic drugs for AAA. We aimed to identify novel plasma biomarkers or potential therapeutic targets for AAA using a high-throughput protein array-based method. Proteomics expression profiles were investigated in plasma from AAA patients and healthy controls (HC) using 440-cytokine protein array analysis. Several promising biomarkers were further validated in independent cohorts using enzyme-linked immunosorbent assay (ELISA). Thirty-nine differentially expressed plasma proteins were identified between AAA and HC. Legumain (LGMN) was significantly higher in AAA patients and was validated in another large cohort. Additionally, “AAA without diabetes” (AAN) patients and “AAA complicated with type 2 diabetes mellitus” (AAM) patients had different cytokine expression patterns in their plasma, and nine plasma proteins were differentially expressed among the AAN, AAM, and HC subjects. Delta-like protein 1 (DLL1), receptor tyrosine-protein kinase erbB-3 (ERBB3), and dipeptidyl peptidase 4 (DPPIV) were significantly higher in AAM than in AAN. This study identified several promising plasma biomarkers of AAA. Their role as therapeutic targets for AAA warrants further investigation. Full article
(This article belongs to the Special Issue Biomarkers for Vascular Disease)
Show Figures

Figure 1

Figure 1
<p>Bioinformatics analysis of differentially expressed proteins between AAA patients and healthy controls. (<b>A</b>) Volcano plot of differentially expressed proteins; (<b>B</b>) orthogonal partial least squares discriminant analysis (OPLS−DA) result; (<b>C</b>) disease and biofunction results from IPA analysis; (<b>D</b>) canonical pathway results from IPA analysis.</p>
Full article ">Figure 2
<p>Diagnostic performance of LGMN in the discovery and validation cohorts. (<b>A</b>) ROC analysis of LGMN according to protein array data; (<b>B</b>) ELISA result of LGMN in validation cohort 2; (<b>C</b>) ROC analysis of LGMN in validation cohort 2. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Identification and bioinformatics analyses of differentially expressed proteins between AAM and AAN patients. (<b>A</b>) Principal component analysis (PCA) model between AAM and AAN; (<b>B</b>) Clustering heatmap of differential proteins between the two groups; (<b>C</b>) Disease and biofunction analysis; (<b>D</b>) Canonical pathway analysis.</p>
Full article ">Figure 4
<p>Exploration of crucial proteins among AAM and AAN patients and healthy controls. (<b>A</b>) Protein array results; (<b>B</b>) ELISA results in validation cohort 2. Data represent mean ± standard error. One-way ANOVA followed by multiple comparisons was conducted to examine the differences among the three groups. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns: Not significant. DLL1: delta-like protein 1, DPPIV: dipeptidyl peptidase 4, ERBB3: receptor tyrosine-protein kinase erbB-3.</p>
Full article ">
16 pages, 2599 KiB  
Article
Legumain Activity Is Controlled by Extended Active Site Residues and Substrate Conformation
by Tasneem Elamin, Hans Brandstetter and Elfriede Dall
Int. J. Mol. Sci. 2022, 23(20), 12548; https://doi.org/10.3390/ijms232012548 - 19 Oct 2022
Cited by 3 | Viewed by 2208
Abstract
Legumain is a lysosomal cysteine protease with strict specificity for cleaving after asparagine residues. By sequence comparison, legumain belongs to MEROPS clan CD of the cysteine proteases, which indicates its structural and mechanistic relation to caspases. Contrasting caspases, legumain harbors a pH-dependent ligase [...] Read more.
Legumain is a lysosomal cysteine protease with strict specificity for cleaving after asparagine residues. By sequence comparison, legumain belongs to MEROPS clan CD of the cysteine proteases, which indicates its structural and mechanistic relation to caspases. Contrasting caspases, legumain harbors a pH-dependent ligase activity in addition to the protease activity. Although we already have a significant body of knowledge on the catalytic activities of legumain, many mechanistic details are still elusive. In this study, we provide evidence that extended active site residues and substrate conformation are steering legumain activities. Biochemical experiments and bioinformatics analysis showed that the catalytic Cys189 and His148 residues are regulated by sterically close Glu190, Ser215 and Asn42 residues. While Glu190 serves as an activity brake, Ser215 and Asn42 have a favorable effect on legumain protease activity. Mutagenesis studies using caspase-9 as model enzyme additionally showed that a similar Glu190 activity brake is also implemented in the caspases. Furthermore, we show that the substrate’s conformational flexibility determines whether it will be hydrolyzed or ligated by legumain. The functional understanding of the extended active site residues and of substrate prerequisites will allow us to engineer proteases with increased enzymatic activity and better ligase substrates, with relevance for biotechnological applications. Full article
(This article belongs to the Special Issue Biocatalysis: Mechanisms of Proteolytic Enzymes 2.0)
Show Figures

Figure 1

Figure 1
<p>Structure-based sequence alignment of clan CD proteases revealed strict conservation of catalytic cysteine and histidine residues and protease specific regulatory residues. The catalytic Cys189 and His148 (human legumain numbering) are indicated by a grey box. Regulatory residues are indicated by blue boxes. The alignment was prepared based on structures of the following clan CD proteases: human legumain (pdb 7O50), mouse legumain (pdb 4NOJ), jack bean legumain (pdb 6XT5), butelase-1 (pdb 6DHI), PIGK (phosphatidylinositol glycan anchor biosynthesis class K protein, pdb 7W72), caspase-1 (pdb 1ICE), caspase-2 (pdb 1PYO), caspase-3 (pdb 1CP3), caspase-9 (pdb 1NW9), yeast metacaspase-1 (YCA1, pdb 4F6O), <span class="html-italic">T. brucei</span> metacaspase-2 (pdb 4AFP), <span class="html-italic">A. thaliana</span> metacaspase-4 (pdb 6W8R), human paracaspase MALT1 (pdb 3UOA), <span class="html-italic">P. gingivalis</span> gingipain K (pdb 4RBM), <span class="html-italic">P. gingivalis</span> gingipain R (pdb 1CVR), <span class="html-italic">V. cholerae</span> RTX cysteine protease (pdb 3EEB), <span class="html-italic">C. difficile</span> TcdB cysteine protease (pdb 3PEE), <span class="html-italic">P. merdae</span> clostripain (pdb 4YEC) and human separase (pdb 7NJ1). Furthermore, sequences of the peptide asparaginyl ligase 2 and 3 from <span class="html-italic">Viola philippica</span> (VpPAL2 and VpPAL3) were included.</p>
Full article ">Figure 2
<p>Asn42 enhances legumain protease activity. (<b>a</b>) Zoom-in view on the active site of legumain in complex with the Ac-Gly-Ser-Asn peptide (GSN peptide; pdb 7o50). Active site residues are shown in grey sticks, the peptide ligand in orange sticks. Please note that the side chain of Cys189 adopted two alternative conformations within this structure. (<b>b</b>) Superposition of the active sites of legumain (grey, pdb 7o50) and <span class="html-italic">P. gingivalis</span> gingipain K (blue, pdb 4rbm). (<b>c</b>) Superposition of the active site of legumain (grey) and caspase-9 (pink, pdb 1jxq). Active site residues are shown in sticks. The P1-Asn residue (grey sticks) of the GSN peptide indicates the position of the S1 specificity pocket. (<b>d</b>) Activity of wild-type, N42A- and N42D-legumain (LEG) measured as turnover of the Bz-Asn-pNA substrate at pH 4.0 and pH 5.5. (<b>e</b>) Thermal unfolding of wild-type (LEG), N42A- and N42D-legumain (LEG-N42A and LEG-N42D), measured at pH 6.0. (<b>f</b>,<b>g</b>) Incubation of legumain with cystatin M/E (hCE; molar ratio 3:1) at pH 4 led to processing after the P1-Asn39 residue. The C-terminal cleavage product hCE(S40-M120) is indicated. Subsequent incubation at pH 7.0 or addition of S-methyl methanethiosulfonate (MMTS) led to resynthesis of the intact cystatin M/E by re-ligation of the Asn39-Ser40 peptide bond. MMTS leads to a covalent thiomethylation of the catalytic Cys189. (<b>h</b>,<b>i</b>) Bands corresponding to intact cystatin M/E and to the C-terminal cleavage product hCE(S40-M120) shown in (<b>f</b>,<b>g</b>) were quantified using ImageJ.</p>
Full article ">Figure 3
<p>A pKa tuning switch is also implemented in caspases. (<b>a</b>) Zoom-in view on the active site of caspase-9 (pdb 1jxq). Active site residues are shown in pink sticks, the Z-VAD-fmk inhibitor is shown in blue sticks. (<b>b</b>) Sequence alignment of human caspases. The catalytic Cys285 residue is indicated by a red star. The pKa switch at position 288 is highlighted by a blue box. Uniprot IDs are specified in the material and methods section. (<b>c</b>) Activity of ΔCARD-caspase-9 and the ΔCARD-caspase-9-E288K mutant measured as turnover of the VAD-AMC substrate. (<b>d</b>) K<sub>M</sub> values of ΔCARD-caspase-9 and the ΔCARD-caspase-9-E288K mutant determined using the VAD-AMC substrate.</p>
Full article ">Figure 4
<p>Ser215 is enhancing legumain protease activity towards peptidic substrates. (<b>a</b>) Sequence alignment of human, mouse, jack bean, <span class="html-italic">O. sativa</span> (rice), and <span class="html-italic">H. annuus</span> (sunflower) legumain and the PIGK protein (phosphatidylinositol glycan anchor biosynthesis class K protein, part of the glycosylphosphatidylinositol transamidase complex). Residue 215 (human legumain numbering) is highlighted in red. Uniprot IDs of the sequences used are specified in the material and methods section. (<b>b</b>) Relative activity of wild-type legumain (LEG), and the legumain-S215N and -S215D mutants measured as turnover of the AAN-AMC substrate at pH 5.5. (<b>c</b>) Thermal unfolding of wild-type (LEG), S215A- and S215D-legumain variants (LEG-S215A and LEG-S215D), measured at pH 6.0. (<b>d</b>) Incubation of legumain with cystatin M/E (hCE) at pH 4.0 led to processing of cystatin M/E after the P1-Asn39 residue. The C-terminal cleavage product hCE(S40-M120) is indicated. Subsequent incubation at pH 7.0 or addition of S-methyl methanethiosulfonate (MMTS) led to resynthesis of the intact cystatin M/E by re-ligation of the Asn39-Ser40 peptide bond. MMTS leads to a covalent thiomethylation of the catalytic Cys189. (<b>e</b>) Bands corresponding to intact cystatin M/E and to the C-terminal cleavage product hCE(S40-M120) shown in (<b>d</b>) were quantified using ImageJ.</p>
Full article ">Figure 5
<p>Substrate conformation is critical for hydrolysis and ligation. (<b>a</b>) Structure of legumain (LEG, green) in complex with cystatin M/E (hCE, blue, pdb 4n6o). The reactive center loop (RCL) is shown in orange, the legumain exosite loop (LEL) in purple. (<b>b</b>) Zoom-in view on the RCL (orange). The catalytic Cys189 is indicated in green sticks, the P2–P1′ residues on the RCL as orange sticks and Lys75 on the LEL as purple sticks. (<b>c</b>) Sequence alignment of family 2 cystatins C, D, M/E, F, S, SA, and SN. Lys75 is highlighted in purple. (<b>d</b>) Residual activity of legumain measured as turnover of the AAN-AMC substrate upon incubation with wild-type cystatin M/E (hCE-wt) or the cystatin M/E-K75A (hCE-K75A) mutant at pH 5.5. (<b>e</b>) SDS-PAGE gel after incubating legumain with wild-type cystatin M/E at indicated pH values for 1 h at 37 °C. (<b>f</b>) Same as (<b>e</b>) but using the cystatin M/E-K75A mutant. The C-terminal cleavage product hCE(S40-M120) is indicated. (<b>g</b>) Incubation of legumain with a 4-fold molar excess of cystatin M/E or the cystatin M/E-K75A mutant at pH 4.0 led to processing of the cystatin after the P1-Asn39 residue. Subsequent incubation at pH 7.0 or addition of S-methyl methanethiosulfonate (MMTS) led to re-ligation of the Asn39-Ser40 peptide bond in wild-type cystatin M/E but not in the cystatin M/E-K75A mutant. (<b>h</b>) Legumain was co-incubated with the cystatin M/E-K75A mutant at pH 4.0 in a 1:4 molar ratio and subjected to size exclusion chromatography (SEC) at pH 4.0. In control experiments legumain alone (LEG) or cystatin M/E-K75A alone were loaded onto the SEC column. (<b>i</b>) Same as (<b>h</b>) but legumain was pre-incubated with MMTS to covalently block the catalytic Cys189 and thereby prevent cleavage of the cystatin. Indicated peak fractions of the experiment containing both legumain and cystatin M/E-K75A were analyzed by SDS-PAGE.</p>
Full article ">Figure 6
<p>Legumain activities are regulated by extended active site residues and substrate conformation. Zoom-in view on the active site of legumain as seen in the crystal structure of legumain in complex with cystatin M/E (pdb 4n6o). The catalytic Cys189 and His148 residues are shown in green sticks, regulatory residues on legumain in blue sticks. The position of the substrate is indicated by the reactive center loop of cystatin M/E (orange).</p>
Full article ">
21 pages, 3182 KiB  
Article
Fetal Programming of the Endocrine Pancreas: Impact of a Maternal Low-Protein Diet on Gene Expression in the Perinatal Rat Pancreas
by Louise Winkel, Morten Rasmussen, Louise Larsen, Louise T. Dalgaard and Jens H. Nielsen
Int. J. Mol. Sci. 2022, 23(19), 11057; https://doi.org/10.3390/ijms231911057 - 21 Sep 2022
Viewed by 2301
Abstract
In rats, the time of birth is characterized by a transient rise in beta cell replication, as well as beta cell neogenesis and the functional maturation of the endocrine pancreas. However, the knowledge of the gene expression during this period of beta cell [...] Read more.
In rats, the time of birth is characterized by a transient rise in beta cell replication, as well as beta cell neogenesis and the functional maturation of the endocrine pancreas. However, the knowledge of the gene expression during this period of beta cell expansion is incomplete. The aim was to characterize the perinatal rat pancreas transcriptome and to identify regulatory pathways differentially regulated at the whole organ level in the offspring of mothers fed a regular control diet (CO) and of mothers fed a low-protein diet (LP). We performed mRNA expression profiling via the microarray analysis of total rat pancreas samples at embryonic day (E) 20 and postnatal days (P) 0 and 2. In the CO group, pancreas metabolic pathways related to sterol and lipid metabolism were highly enriched, whereas the LP diet induced changes in transcripts involved in RNA transcription and gene regulation, as well as cell migration and apoptosis. Moreover, a number of individual transcripts were markedly upregulated at P0 in the CO pancreas: growth arrest specific 6 (Gas6), legumain (Lgmn), Ets variant gene 5 (Etv5), alpha-fetoprotein (Afp), dual-specificity phosphatase 6 (Dusp6), and angiopoietin-like 4 (Angptl4). The LP diet induced the downregulation of a large number of transcripts, including neurogenin 3 (Neurog3), Etv5, Gas6, Dusp6, signaling transducer and activator of transcription 3 (Stat3), growth hormone receptor (Ghr), prolactin receptor (Prlr), and Gas6 receptor (AXL receptor tyrosine kinase; Axl), whereas upregulated transcripts were related to inflammatory responses and cell motility. We identified differentially regulated genes and transcriptional networks in the perinatal pancreas. These data revealed marked adaptations of exocrine and endocrine in the pancreas to the low-protein diet, and the data can contribute to identifying novel regulators of beta cell mass expansion and functional maturation and may provide a valuable tool in the generation of fully functional beta cells from stem cells to be used in replacement therapy. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Comparison of fold changes (FCs) in mRNA expression levels between P0 and E20 as obtained by hybridization to Affymetrix GeneChip versus RTqPCR for the number of selected up- and downregulated genes. There are no error bars, since data are presented as fold changes. Note: Pnlip: pancreatic lipase; Bcat2: branched-chain amino acid transaminase 2; Gas6: growth-arrest-specific 6; Afp: alpha-fetoprotein; Ngn3: neurog3; Srebf1/2: sterol regulatory element binding protein 1/2; Ins2: insulin 2; Pdx1: pancreatic duodenal homeobox 1; Egr1: early growth response factor 1. (<b>B</b>) Dendrogram for clustering using centered correlation and average linkage. Samples from P0 and P2 cluster closer together, indicating a more similar profile compared to samples from E20. (<b>C</b>) Heat map generated from the dCHIP hierarchical cluster analysis. Note: E20: embryonic day 20; P0: day 0 postpartum; P2: day two postpartum. Blue: Low expression. Red: High expression.</p>
Full article ">Figure 2
<p>Expression and cellular localization of <span class="html-italic">Afp</span> in rat pancreas samples at E20, P0, and P2. (<b>A</b>,<b>C</b>,<b>E</b>) In situ hybridization of <span class="html-italic">Afp</span> mRNA using a dioxigenin-labeled LNA™ probe targeted against the same rat <span class="html-italic">Afp</span> sequence as the primers for RTqPCR (Qiagen, Exiqon, Vedbaek, Denmark) at E20 (<b>A</b>), P0 (<b>C</b>), and P2 (<b>E</b>). <span class="html-italic">Afp</span> is primarily expressed in association with islets of Langerhans in vessels and intra-islet capillaries. (<b>B</b>,<b>D</b>,<b>F</b>) Immunohistochemical detection (200× magnification) of Afp protein in 3 µm tissue sections from rat pancreas samples at E20 (<b>B</b>), P0 (<b>D</b>), and P2 (<b>F</b>). Afp is primarily detected in association with the endocrine compartments or ducts (arrows). Inserts at 400× magnification. (<b>G</b>) RTqPCR of <span class="html-italic">Afp</span> mRNA in rat pancreas samples at E20, P0, and P2. The mRNA values are normalized to <span class="html-italic">Rpl13alpha</span>. <span class="html-italic">Afp</span> mRNA expression is significantly increased at P0 compared to E20 and P2 (two-tailed <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.01). Note: *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Functionally annotated gene clusters in normal perinatal rat pancreas samples. Differentially regulated transcripts in perinatal rat pancreas were clustered according to expression profiles over time and analyzed for enriched functional annotations using DAVID Functional Annotation Clustering. The most significantly enriched biological process in the highest scoring cluster is presented above each graph. Data points represent mean intensities of the associated gene cluster at the given time point. (<b>A</b>) Lipid metabolic process, (<b>B</b>) proteolysis, (<b>C</b>) activation of the immune system, (<b>D</b>) cell development, (<b>E</b>) sterol biosynthetic process, (<b>F</b>) alcohol metabolic process. Error bars (SE) are included to illustrate the variability in expression levels. <span class="html-italic">p</span>-value: modified one-tailed Fisher’s exact probability value (EASE score); E-score: enrichment score, the geometric mean (in -log scale) of a member’s <span class="html-italic">p</span>-values (EASE scores) in the corresponding annotation cluster.</p>
Full article ">Figure 4
<p>Relative <span class="html-italic">Ins2, Pdx-1, Neurog3, Ghr,</span> and <span class="html-italic">Prlr</span> mRNA expression in perinatal rat pancreas from control (Ctrl) and low-protein (LP) diet animals. Messenger RNA levels measured using RTqPCR in whole rat pancreas samples at days E20, P0, and P2. Values are normalized to levels at E20 within each data set, <span class="html-italic">n</span> = 3–6. Data are given as means + SEM. (<b>A</b>) <span class="html-italic">Ins2</span>, (<b>B</b>) <span class="html-italic">Pdx1</span>, (<b>C</b>) <span class="html-italic">Neurog3</span>, (<b>D</b>) <span class="html-italic">Ghr</span>, (<b>E</b>) <span class="html-italic">Prlr</span>, and (<b>F</b>) immunohistochemical stainings for Neurog3 in the perinatal rat pancreas at P0 (200× magnification). Data in panels (<b>A</b>–<b>E</b>) were tested using 2-way ANOVA with Bonferroni post tests for control vs. LP animals. Asterisks indicate significance levels of individual comparisons. Note: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Gene Ontology biological processes and molecular functions,) which are significantly over-represented in the gene lists of differentially expressed genes at E20 vs. P0, and significantly enriched transcription factor binding sites (TFBS) based on differentially regulated genes between P0 and P2.</p>
Full article ">Figure 6
<p>Cluster analysis of genes regulated by gestational low-protein diet. (<b>A</b>,<b>B</b>) Venn diagrams based on lists of upregulated (<b>A</b>) and downregulated (<b>B</b>) genes in the perinatal pancreas (E20, P0, and P2) programmed by the low-protein diet during gestation. (<b>C</b>) Heat map and cluster analyses of low-protein diet-regulated genes in the perinatal period. LP: Low-protein; CO: control. Green: Upregulated and Red: Down regulated.</p>
Full article ">Figure 7
<p>Gene Ontology biological processes (GO:BP) and molecular functions (GO:MF), which are significantly over-represented in the gene lists of downregulated genes caused by the low-protein diet, and significantly enriched transcription factor binding sites (TFBS) based on downregulated genes caused by the low-protein diet during gestation.</p>
Full article ">
21 pages, 7658 KiB  
Article
Roles of Species-Specific Legumains in Pathogenicity of the Pinewood Nematode Bursaphelenchus xylophilus
by Xi Zhang, Runmao Lin, Jian Ling, Yunsheng Wang, Feifei Qin, Junru Lu, Xin Sun, Manling Zou, Jing Qi, Bingyan Xie and Xinyue Cheng
Int. J. Mol. Sci. 2022, 23(18), 10437; https://doi.org/10.3390/ijms231810437 - 9 Sep 2022
Cited by 2 | Viewed by 1743
Abstract
Peptidases are very important to parasites, which have central roles in parasite biology and pathogenesis. In this study, by comparative genome analysis, genome-wide peptidase diversities among plant-parasitic nematodes are estimated. We find that genes encoding cysteine peptidases in family C13 (legumain) are significantly [...] Read more.
Peptidases are very important to parasites, which have central roles in parasite biology and pathogenesis. In this study, by comparative genome analysis, genome-wide peptidase diversities among plant-parasitic nematodes are estimated. We find that genes encoding cysteine peptidases in family C13 (legumain) are significantly abundant in pine wood nematodes Bursaphelenchus genomes, compared to those in other plant-parasitic nematodes. By phylogenetic analysis, a clade of B. xylophilus-specific legumain is identified. RT-qPCR detection shows that these genes are highly expressed at early stage during the nematode infection process. Utilizing transgene technology, cDNAs of three species-specific legumain were introduced into the Arabidopsis γvpe mutant. Functional complementation assay shows that these B. xylophilus legumains can fully complement the activity of Arabidopsis γVPE to mediate plant cell death triggered by the fungal toxin FB1. Secretory activities of these legumains are experimentally validated. By comparative transcriptome analysis, genes involved in plant cell death mediated by legumains are identified, which enrich in GO terms related to ubiquitin protein transferase activity in category molecular function, and response to stimuli in category biological process. Our results suggest that B. xylophilu-specific legumains have potential as effectors to be involved in nematode-plant interaction and can be related to host cell death. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Predicted genes encoding peptidases in plant-parasitic nematode genomes: (<b>a</b>) heatmap of peptidases distribution. The red dot marks the peptidase C13 family (legumains); (<b>b</b>) number of peptidases in each genome.</p>
Full article ">Figure 2
<p>Phylogenetic relationship of legumains in plant-parasitic nematodes and <span class="html-italic">C. elegans</span>: (<b>a</b>) phylogeny of nematode legumains and their primary structures, i.e., a peptidase_C13 domain (PF01650); and (<b>a</b>) C-terminal prodomain of legumain (legumain_C, cd2115). The phylogenetic tree was constructed based on full-length protein sequences. Bootstrap values of ML/NJ are showed on branches, and bootstrap value less than 50% is replaced by the marker --. The <span class="html-italic">B. xylophilus</span>-specific legumains are enclosed in the red box; (<b>b</b>) putative duplications of legumain-coding genes in the <span class="html-italic">B. xylophilus</span> genome (BxCN).</p>
Full article ">Figure 3
<p>Structural comparison of legumains: (<b>a</b>) primary protein structural organization of <span class="html-italic">B. xylophilus</span>-specific legumain (BxCN10334), <span class="html-italic">Arabidopsis</span> γVPE (AT4G32940), and human AEP (NP_001008530). They have a signal peptide (gray box) at the N-terminus, a peptidase_C13 domain (PF01650) (blue box) and a C-terminal prodomain of legumain (legumain_C, cd2115) (orange box). The C13 domain includes four essential amino acid residues (R, H, C, S), which form the substrate pocket, and the middle two (H, C) are catalytic dyad; (<b>b</b>) prediction of spatial structures of <span class="html-italic">B. xylophilus</span>-specific legumain, <span class="html-italic">Arabidopsis</span> γVPE, and human AEP, with RoseTTAFold online (job IDs 207107, 207108 and 207109, respectively). Pink: signal peptide; blue: peptidase C13 family domain; yellow: C-terminal of prodomain of legumain; red: the essential amino acid residues; (<b>c</b>) protein structure comparison by TM-align, between <span class="html-italic">B. xylophilus</span> legumain and <span class="html-italic">Arabidopsis</span> γVPE, <span class="html-italic">B. xylophilus</span> legumain and human AEP. Pink represent <span class="html-italic">B. xylophilus</span> legumain, blue represent <span class="html-italic">Arabidopsis</span> γVPE or human AEP.</p>
Full article ">Figure 4
<p>Expression patterns and functional complementation assay of <span class="html-italic">B. xylophilus</span>–specific legumains: (<b>a</b>) RT-qPCR detection of mRNA expression of <span class="html-italic">B. xylophilus</span>-specific legumains after the nematode was inoculated on five years old pine seedlings (<span class="html-italic">P. thunbergii</span>) for 1 to 15 days, taking 0 d (1–2 h) as control. The EF-1α gene was used as an internal control. Bars are standard errors (SE); (<b>b</b>) two BxCN legumain genes (BxCN10334, BxCN10337) were introduced into the <span class="html-italic">Arabidopsis Δγvpe</span> mutant (Salk_024036C). Necrotic spots are observed on leaves of homozygous T3 plants after infiltration with FB1 (10 mM, in 0.1% methanol) for five days, which are similar to those in the wild type <span class="html-italic">Arabidopsis</span> plants infiltration with FB1. The mutant plants infiltrated with FB1 and the wild type plants infiltrated with methanol (solvent) are taken as controls; (<b>c</b>) quantitative assay of complementation effects of transgenic lines with four <span class="html-italic">B. xylophilus</span>-specific legumains and a pine VPE (<span class="html-italic">Pinus taeda</span> PITA_000069534), by infiltration with 10 μL of FB1 (in 0.1% methanol) on each leaf. Necrotic lesions on <span class="html-italic">Δγvpe</span> mutant and the wild type <span class="html-italic">Arabidopsis</span> plants are taken as controls; (<b>d</b>) comparison of the mean diameters of necrotic spots in different transgenic lines and the wild-type <span class="html-italic">Arabidopsis</span> plants, compared with the <span class="html-italic">Δγvpe</span> mutant. Black dots represent outliers.</p>
Full article ">Figure 5
<p>Localization of three BxCN legumain transcripts in the nematode by in situ hybridization: (<b>a</b>) hybridization with digoxigenin-labelled cDNA probes. Scale bar, 50 μm; (<b>b</b>) hybridization with 5′- end FITC-labelled cDNA probes. DG, dorsal glands; M, median bulb; S, stylet; SVG, subventral glands. Scale bar, 20 μm.</p>
Full article ">Figure 6
<p>Yeast invertase secretion assay of the signal peptides of the above three BxCN legumains. Yeast YTK12 strain carrying signal peptides of BxCN10334, BxCN10337, and BxCN10284, are able to grow in raffinose-containing YPRAA medium, and react with TTC to display red color. YTK12 carrying the Avrblb2 and the pSUC2 vector are used as positive and negative controls, respectively. CMD-W, YPDA, and YPRAA, are culture media. TTC, 2,3,5-triphenyltetrazolium chloride.</p>
Full article ">Figure 7
<p>Differentially expressed genes (DEGs) in the four types of <span class="html-italic">Arabidopsis</span> transcriptomes and enrichment in GO terms: (<b>a</b>) Venn diagram shows common and specific DEGs among the transcriptomes of the wild-type and the two transgenic lines introduced with <span class="html-italic">B. xylophilus</span>-specific legumain-coding genes (BxCN10334 and BxCN10337), each compared with the control (<span class="html-italic">∆γvpe</span> mutant); (<b>b</b>) enriched GO terms of the 59 common DEGs, which with identical expression patterns among the above three transcriptomes; (<b>c</b>) the top enriched GO terms of additional 1333 DEGs, which have identical expression patterns between the two transgenic lines transcriptomes.</p>
Full article ">
Back to TopTop