Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Sci Rep ; 13(1): 22255, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38097707

ABSTRACT

Cisplatin (CP) induces acute kidney injury (AKI) whereby proximal tubules undergo regulated necrosis. Repair is almost complete after a single dose. We now demonstrate a role for Apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (Apobec-1) that is prominently expressed at the interface between acute and chronic kidney injury (CKD), in the recovery from AKI. Apobec-1 knockout (KO) mice exhibited greater mortality than in wild type (WT) and more severe AKI in both CP- and unilateral ischemia reperfusion (IR) with nephrectomy. Specifically, plasma creatinine (pCr) 2.6 ± 0.70 mg/dL for KO, n = 10 and 0.16 ± 0.02 for WT, n = 6, p < 0.0001 in CP model and 1.34 ± 0.22 mg/dL vs 0.75 ± 0.06, n = 5, p < 0.05 in IR model. The kidneys of Apobec-1 KO mice showed increased necrosis, increased expression of KIM-1, NGAL, RIPK1, ASCL4 and increased lipid accumulation compared to WT kidneys (p < 0.01). Neutrophils and activated T cells were both increased, while macrophages were reduced in kidneys of Apobec-1 KO animals. Overexpression of Apobec-1 in mouse proximal tubule cells protected against CP-induced cytotoxicity. These findings suggest that Apobec-1 mediates critical pro-survival responses to renal injury and increasing Apobec-1 expression could be an effective strategy to mitigate AKI.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Mice , Animals , APOBEC-1 Deaminase/metabolism , Cisplatin/adverse effects , Cisplatin/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Kidney/metabolism , Necrosis/metabolism , Mice, Knockout , Reperfusion Injury/metabolism , Mice, Inbred C57BL
2.
Eur J Cancer ; 165: 81-96, 2022 04.
Article in English | MEDLINE | ID: mdl-35219026

ABSTRACT

BACKGROUND: Although programmed cell death protein 1 (PD-1) inhibitors have revolutionised treatment for advanced melanoma, not all patients respond. We previously showed that inhibition of the flavoprotein renalase (RNLS) in preclinical melanoma models decreases tumour growth. We hypothesised that RNLS inhibition promotes tumour rejection by effects on the tumour microenvironment (TME). METHODS: We used two distinct murine melanoma models, studied in RNLS knockout (KO) or wild-type (WT) mice. WT mice were treated with the anti-RNLS antibody, m28, with or without anti-PD-1. 10X single-cell RNA-sequencing was used to identify transcriptional differences between treatment groups, and tumour cell content was interrogated by flow cytometry. Samples from patients treated with immunotherapy were examined for RNLS expression by quantitative immunofluorescence. RESULTS: RNLS KO mice injected with wild-type melanoma cells reject their tumours, supporting the importance of RNLS in cells in the TME. This effect was blunted by anti-cluster of differentiation 3. However, MØ-specific RNLS ablation was insufficient to abrogate tumour formation. Anti-RNLS antibody treatment of melanoma-bearing mice resulted in enhanced T cell infiltration and activation and resulted in immune memory on rechallenging mice with injection of melanoma cells. At the single-cell level, treatment with anti-RNLS antibodies resulted in increased tumour density of MØ, neutrophils and lymphocytes and increased expression of IFNγ and granzyme B in natural killer cells and T cells. Intratumoural Forkhead Box P3 + CD4 cells were decreased. In two distinct murine melanoma models, we showed that melanoma-bearing mice treated with anti-RNLS antibodies plus anti-PD-1 had superior tumour shrinkage and survival than with either treatment alone. Importantly, in pretreatment samples from patients treated with PD-1 inhibitors, high RNLS expression was associated with decreased survival (log-rank P = 0.006), independent of other prognostic variables. CONCLUSIONS: RNLS KO results in melanoma tumour regression in a T-cell-dependent fashion. Anti-RNLS antibodies enhance anti-PD-1 activity in two distinct aggressive murine melanoma models resistant to PD-1 inhibitors, supporting the development of anti-RNLS antibodies with PD-1 inhibitors as a novel approach for melanomas poorly responsive to anti-PD-1.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Animals , Humans , Immunotherapy , Melanoma/drug therapy , Mice , Monoamine Oxidase/therapeutic use , Tumor Microenvironment
3.
J Am Soc Nephrol ; 33(2): 342-356, 2022 02.
Article in English | MEDLINE | ID: mdl-34921111

ABSTRACT

BACKGROUND: Repeated administration of cisplatin causes CKD. In previous studies, we reported that the kidney-secreted survival protein renalase (RNLS) and an agonist peptide protected mice from cisplatin-induced AKI. METHODS: To investigate whether kidney-targeted delivery of RNLS might prevent cisplatin-induced CKD in a mouse model, we achieved specific delivery of a RNLS agonist peptide (RP81) to the renal proximal tubule by encapsulating the peptide in mesoscale nanoparticles (MNPs). We used genetic deletion of RNLS, single-cell RNA sequencing analysis, and Western blotting to determine efficacy and to explore underlying mechanisms. We also measured plasma RNLS in patients with advanced head and neck squamous cell carcinoma receiving their first dose of cisplatin chemotherapy. RESULTS: In mice with CKD induced by cisplatin, we observed an approximate 60% reduction of kidney RNLS; genetic deletion of RNLS was associated with significantly more severe cisplatin-induced CKD. In this severe model of cisplatin-induced CKD, systemic administration of MNP-encapsulated RP81 (RP81-MNP) significantly reduced CKD as assessed by plasma creatinine and histology. It also decreased inflammatory cytokines in plasma and inhibited regulated necrosis in kidney. Single-cell RNA sequencing analyses revealed that RP81-MNP preserved epithelial components of the nephron and the vasculature and suppressed inflammatory macrophages and myofibroblasts. In patients receiving their first dose of cisplatin chemotherapy, plasma RNLS levels trended lower at day 14 post-treatment. CONCLUSIONS: Kidney-targeted delivery of RNLS agonist RP81-MNP protects against cisplatin-induced CKD by decreasing cell death and improving the viability of the renal proximal tubule. These findings suggest that such an approach might mitigate the development of CKD in patients receiving cisplatin cancer chemotherapy.


Subject(s)
Cisplatin/adverse effects , Monoamine Oxidase/metabolism , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/prevention & control , Amino Acid Sequence , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cell Line , Cisplatin/administration & dosage , Creatinine/blood , Disease Models, Animal , Gene Expression/drug effects , Glomerular Filtration Rate , Hepatitis A Virus Cellular Receptor 1/blood , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoamine Oxidase/deficiency , Monoamine Oxidase/genetics , Nanocapsules/administration & dosage , Peptides/administration & dosage , Peptides/genetics , Renal Insufficiency, Chronic/pathology
4.
PLoS One ; 16(9): e0250539, 2021.
Article in English | MEDLINE | ID: mdl-34587190

ABSTRACT

Dysregulated expression of the secretory protein renalase can promote pancreatic ductal adenocarcinoma (PDAC) growth in animal models. We characterized renalase expression in premalignant and malignant PDAC tissue and investigated whether plasma renalase levels corresponded to clinical PDAC characteristics. Renalase immunohistochemistry was used to determine the presence and distribution of renalase in normal pancreas, chronic pancreatitis, PDAC precursor lesions, and PDAC tissues. Associations between pretreatment plasma renalase and PDAC clinical status were assessed in patients with varied clinical stages of PDAC and included tumor characteristics, surgical resection in locally advanced/borderline resectable PDAC, and overall survival. Data were retrospectively obtained and correlated using non-parametric analysis. Little to no renalase was detected by histochemistry in the normal pancreatic head in the absence of abdominal trauma. In chronic pancreatitis, renalase immunoreactivity localized to peri-acinar spindle-shaped cells in some samples. It was also widely present in PDAC precursor lesions and PDAC tissue. Among 240 patients with PDAC, elevated plasma renalase levels were associated with worse tumor characteristics, including greater angiolymphatic invasion (80.0% vs. 58.1%, p = 0.012) and greater node positive disease (76.5% vs. 56.5%, p = 0.024). Overall survival was worse in patients with high plasma renalase levels with median follow-up of 27.70 months vs. 65.03 months (p < 0.001). Renalase levels also predicted whether patients with locally advanced/borderline resectable PDAC underwent resection (AUC 0.674; 95%CI 0.42-0.82, p = 0.04). Overall tissue renalase was increased in both premalignant and malignant PDAC tissues compared to normal pancreas. Elevated plasma renalase levels were associated with advanced tumor characteristics, decreased overall survival, and reduced resectability in patients with locally advanced/borderline resectable PDAC. These studies show that renalase levels are increased in premalignant pancreatic tissues and that its levels in plasma correspond to the clinical behavior of PDAC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Pancreatic Ductal/pathology , Monoamine Oxidase/blood , Pancreatic Neoplasms/pathology , Up-Regulation , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/mortality , Case-Control Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Grading , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/mortality , Prognosis , Prospective Studies , Retrospective Studies , Survival Analysis , Young Adult , Pancreatic Neoplasms
5.
J Cell Mol Med ; 21(7): 1260-1265, 2017 07.
Article in English | MEDLINE | ID: mdl-28238213

ABSTRACT

Renalase was discovered as a protein synthesized by the kidney and secreted in blood where it circulates at a concentration of approximately 3-5 µg/ml. Initial reports suggested that it functioned as an NAD(P)H oxidase and could oxidize catecholamines. Administration of renalase lowers blood pressure and heart rate and also protects cells and organs against ischaemic and toxic injury. Although renalase's protective effect was initially ascribed to its oxidase properties, a paradigm shift in our understanding of the cellular actions of renalase is underway. We now understand that, independent of its enzymatic properties, renalase functions as a cytokine that provides protection to cells, tissues and organs by interacting with its receptor to activate protein kinase B, JAK/STAT, and the mitogen-activated protein kinase pathways. In addition, recent studies suggest that dysregulated renalase signalling may promote survival of several tumour cells due to its capacity to augment expression of growth-related genes. In this review, we focus on the cytoprotective actions of renalase and its capacity to sustain cancer cell growth and also the translational opportunities these findings represent for the development of novel therapeutic strategies for organ injury and cancer.


Subject(s)
Cytokines/metabolism , Kidney/metabolism , Monoamine Oxidase/metabolism , Neoplasms/drug therapy , Blood Pressure/drug effects , Catecholamines/metabolism , Cytokines/genetics , Heart Rate/drug effects , Humans , Kidney/enzymology , Kidney/pathology , Monoamine Oxidase/therapeutic use , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Oxidation-Reduction
6.
Nucleic Acids Res ; 41(8): 4650-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23449220

ABSTRACT

Therapeutic small interfering RNAs (siRNAs) are composed of chemically modified nucleotides, which enhance RNA stability and increase affinity in Watson-Crick base pairing. However, the precise fate of such modified nucleotides once the siRNA is degraded within the cell is unknown. Previously, we demonstrated that deoxythymidine release from degraded siRNAs reversed the cytotoxicity of thymidylate synthase (TS)-targeted siRNAs and other TS inhibitor compounds. We hypothesized that siRNAs could be designed with specific nucleoside analogues that, once released, would enhance siRNA cytotoxicity. TS-targeted siRNAs were designed that contained 5-fluoro-2'-deoxyuridine (FdU) moieties at various locations within the siRNA. After transfection, these siRNAs suppressed TS protein and messenger RNA expression with different efficiencies depending on the location of the FdU modification. FdU was rapidly released from the siRNA as evidenced by formation of the covalent inhibitory ternary complex formed between TS protein and the FdU metabolite, FdUMP. These modified siRNAs exhibited 10-100-fold greater cytotoxicity and induced multiple DNA damage repair and apoptotic pathways when compared with control siRNAs. The strategy of designing siRNA molecules that incorporate cytotoxic nucleosides represents a potentially novel drug development approach for the treatment of cancer and other human diseases.


Subject(s)
Deoxyuridine/analogs & derivatives , RNA, Small Interfering/chemistry , RNA, Small Interfering/toxicity , Apoptosis , Cell Line, Tumor , Deoxyuridine/toxicity , Humans , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Transfection
7.
J Laparoendosc Adv Surg Tech A ; 19(3): 405-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19215211

ABSTRACT

OBJECTIVES: The aim of this study was to evaluate the effect of a self-releasing biliary stent antegradely placed during laparoscopic common bile duct exploration (LCBDE) for choledocholithiasis. MATERIALS AND METHODS: The soft biliary stent, made of polyurethane, was designed as a J-umbrella form with a pigtailed duodenal part and an umbrella-like biliary anchoring part shaped with the rapidly absorbable suture. After the clearance of stones during LCBDE, a guide wire was inserted into the duodenum through the choledochoscope. The stent was advanced over the guide wire until the pigtail of the stent entered the duodenum. The choledochotomy was primarily closed. RESULTS: This technique has been performed on 33 patients with choledocholithiasis. The median length of postoperative hospital stay was 4.3 (3-8) days. All the stents were eliminated from the bile ducts and discharged out of the body. The median time of the stent stay in the body was 13.6 (+/- 2.55) days. Transient hyperamylasemia occurred in 4 of the 33 (12.1%) patients, and stent occlusion occurred in 1 patient who recovered soon after treatment. No bile leak, biliary infection, or stent dislocation was observed. During the follow-up of 12 months, no biliary infection, residual calculi, or stricture occurred. CONCLUSION: This novel self-releasing stent is safe and effective in the laparoscopic treatment of choledocholithiasis, and the subsequent removal of the stent can be avoided.


Subject(s)
Choledocholithiasis/surgery , Laparoscopy/methods , Stents , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prosthesis Design , Treatment Outcome
8.
Biochem Biophys Res Commun ; 369(3): 795-800, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18045573

ABSTRACT

Human dihydrofolate reductase (DHFR) is a critical target in cancer chemotherapy. Previous studies showed that an 82-nt RNA fragment within the DHFR mRNA protein-coding region functions as a DHFR cis-acting response element. In this study, we further investigated the key elements contained within this sequence that are required for the DHFR mRNA-DHFR protein interaction. Using enzymatic foot-printing assays and RNA-binding experiments, we isolated a 27-nt sequence (DHFR27, corresponding to nts 407-433), which bound with high affinity and specificity to human DHFR to form a ribonucleoprotein complex. In vivo transient transfection experiments using a luciferase reporter system revealed that DHFR27 RNA could repress the luciferase expression in a DHFR-dependent manner when placed upstream of luciferase mRNA. This work provides new insights into the essential molecular elements that mediate RNA-protein interactions.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Regulatory Sequences, Ribonucleic Acid , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Base Sequence , Cell Line, Tumor , Genes, Reporter , Humans , Luciferases, Renilla/genetics , Molecular Sequence Data , RNA, Messenger/chemistry , Tetrahydrofolate Dehydrogenase/chemistry
9.
Front Biosci ; 9: 2521-6, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15353304

ABSTRACT

The folate-dependent enzymes, thymidylate synthase (TS) and dihydrofolate reductase (DHFR) are critical for providing the requisite nucleotide precursors for maintaining DNA synthesis and DNA repair. In addition to their essential roles in enzyme catalysis, these two enzymes have now been shown to function as RNA binding proteins. Using in vitro and in vivo experimental model systems, we have shown that the functional consequence of binding of TS protein to its own cognate mRNA, as well as binding of DHFR to its own DHFR mRNA, is translational repression. Herein, we review and update studies focusing on the translational autoregulatory control of TS and DHFR expression and discuss the molecular elements that are required for these specific RNA-protein interactions. Moreover, we present evidence showing that abrogation of these normal translational autoregulatory feedback mechanisms provides the molecular basis for the rapid development of cellular drug resistance.


Subject(s)
Gene Expression Regulation, Enzymologic , Protein Biosynthesis , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Animals , Cell Line, Tumor , DNA Repair , Drug Resistance , Humans , In Vitro Techniques , Protein Binding , RNA/metabolism
10.
Cancer Res ; 64(4): 1431-5, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14973067

ABSTRACT

RNA interference is a post-transcriptional mechanism by which double-stranded RNA specifically silence expression of a corresponding gene. Small interfering double-stranded RNA (siRNA) of 21-23 nucleotides can induce the process of RNA interference. Studies from our laboratory have shown that translation of thymidylate synthase (TS) mRNA is controlled by its own protein end-product TS in a negative autoregulatory manner. Disruption of this process gives rise to increased synthesis of TS and leads to the development of cellular drug resistance to TS-targeted compounds. As a strategy to inhibit TS expression at the mRNA level, siRNAs were designed to target nucleotides 1058-1077 on human TS mRNA. Transfection of TS1058 siRNA into human colon cancer RKO cells resulted in a dose-dependent inhibition of TS expression with an IC(50) value of 10 pM but had no effect on the expression of alpha-tubulin or topoisomerase I. Inhibition of TS expression by TS1058 was maximal at 48 h and remained suppressed for up to 5 days. Pretreatment of RKO cells with TS1058 siRNA suppressed TS protein induction following exposure to raltitrexed. In addition, TS1058 restored chemosensitivity of the resistant RKO-HTStet cell line to various TS inhibitor compounds. On treatment with TS1058, IC(50) values for raltitrexed, 1843U89, and 5-fluoro-2'-deoxyuridine decreased by approximately 15-16-fold. These studies suggest that TS-targeted siRNAs are effective inhibitors of TS expression and may have therapeutic potential by themselves or as chemosensitizers in combination with TS inhibitor compounds.


Subject(s)
Colonic Neoplasms/drug therapy , Enzyme Inhibitors/therapeutic use , RNA, Double-Stranded/therapeutic use , RNA, Small Interfering/therapeutic use , Thymidylate Synthase/antagonists & inhibitors , Cell Division/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Thymidylate Synthase/genetics
11.
Biochem J ; 378(Pt 3): 999-1006, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14664697

ABSTRACT

Previous studies have shown that human DHFR (dihydrofolate reductase), in addition to its critical role in DNA biosynthesis, functions as an RNA-binding protein. The interaction between DHFR and its own mRNA results in translational repression. In this study, we characterized the cis-acting elements on human DHFR mRNA that are required for the DHFR mRNA-DHFR protein interaction. Using a series of gel-shift and nitrocellulose filter-binding assays, a 164 nt RNA sequence, corresponding to nt 401-564, was identified within the coding region that binds to DHFR protein with an affinity similar to that of full-length DHFR mRNA. To document in vivo biological activity, various DHFR sequences contained within the coding region were cloned on to the 5' end of a luciferase reporter plasmid, and transient transfection experiments were performed using human colon cancer RKO cells. In cells transfected with p644/DHFR:401-564, luciferase activity was decreased by 50% when compared with cells transfected with the p644 plasmid alone. Luciferase mRNA levels were identical under each of these conditions, as determined by Northern-blot analysis. In cells transfected with p644/DHFR:401-564, luciferase activity was restored to almost 100% of control when cells were treated with the antifolate analogue methotrexate or with a short-interfering RNA targeting DHFR mRNA. These findings provide evidence that the DHFR 401-564 sequence is a DHFR-response element. In vitro and in vivo studies further localized this cis-element to an 82 nt sequence corresponding to nt 401-482. This work provides new insights into critical elements that mediate RNA-protein interactions.


Subject(s)
Protein Biosynthesis , RNA, Messenger/chemistry , Regulatory Sequences, Ribonucleic Acid , Response Elements , Tetrahydrofolate Dehydrogenase/genetics , Binding Sites , Cell Line, Tumor , Exons , Gene Expression Regulation , Humans , Luciferases/genetics , Luciferases/metabolism , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/biosynthesis
12.
Biochim Biophys Acta ; 1587(2-3): 174-82, 2002 Jul 18.
Article in English | MEDLINE | ID: mdl-12084459

ABSTRACT

Studies from our laboratory have shown that the folate-dependent enzyme, thymidylate synthase (TS), functions as an RNA binding protein. There is evidence that TS, in addition to interacting with its own TS mRNA, forms a ribonucleoprotein complex with a number of other cellular mRNAs, including those corresponding to the p53 tumor suppressor gene and the myc family of transcription factors. Using both in vitro and in vivo model systems, we have demonstrated that the functional consequence of binding of TS protein to its own cognate mRNA, as well as binding of TS to the p53 mRNA, is translational repression. Herein, we review current work on the translational autoregulatory control of TS expression and discuss the molecular elements that are required for the TS protein-TS mRNA interaction. TS may play a critical role in regulating the cell cycle and the process of apoptosis through its regulatory effects on expression of p53 and perhaps other cell cycle related proteins. Finally, the ability of TS to function as a translational regulator may have important consequences with regard to the development of cellular resistance to various anticancer drugs.


Subject(s)
Gene Expression Regulation , Thymidylate Synthase/genetics , Thymidylate Synthase/metabolism , Animals , Antineoplastic Agents/pharmacology , Genes, p53 , Humans , Models, Biological , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...