Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(4)2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38396999

ABSTRACT

Fibrosis is a chronic pathology resulting from excessive deposition of extracellular matrix components that leads to the loss of tissue function. Pulmonary fibrosis can follow a variety of diverse insults including ischemia, respiratory infection, or exposure to ionizing radiation. Consequently, treatments that attenuate the development of debilitating fibrosis are in desperate need across a range of conditions. Sphingolipid metabolism is a critical regulator of cell proliferation, apoptosis, autophagy, and pathologic inflammation, processes that are all involved in fibrosis. Opaganib (formerly ABC294640) is the first-in-class investigational drug targeting sphingolipid metabolism for the treatment of cancer and inflammatory diseases. Opaganib inhibits key enzymes in sphingolipid metabolism, including sphingosine kinase-2 and dihydroceramide desaturase, thereby reducing inflammation and promoting autophagy. Herein, we demonstrate in mouse models of lung damage following exposure to ionizing radiation that opaganib significantly improved long-term survival associated with reduced lung fibrosis, suppression of granulocyte infiltration, and reduced expression of IL-6 and TNFα at 180 days after radiation. These data further demonstrate that sphingolipid metabolism is a critical regulator of fibrogenesis, and specifically show that opaganib suppresses radiation-induced pulmonary inflammation and fibrosis. Because opaganib has demonstrated an excellent safety profile during clinical testing in other diseases (cancer and COVID-19), the present studies support additional clinical trials with this drug in patients at risk for pulmonary fibrosis.


Subject(s)
Adamantane/analogs & derivatives , Medical Countermeasures , Neoplasms , Pneumonia , Pulmonary Fibrosis , Pyridines , Mice , Animals , Humans , Sphingolipids/metabolism , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology , Fibrosis , Inflammation/drug therapy
2.
Int J Mol Sci ; 23(21)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36361977

ABSTRACT

Exposure to ionizing radiation (IR) is a lingering threat from accidental or terroristic nuclear events, but is also widely used in cancer therapy. In both cases, host inflammatory responses to IR damage normal tissue causing morbidity and possibly mortality to the victim/patient. Opaganib, a first-in-class inhibitor of sphingolipid metabolism, has broad anti-inflammatory and anticancer activity. Opaganib elevates ceramide and reduces sphingosine 1-phosphate (S1P) in cells, conditions that increase the antitumor efficacy of radiation while concomitantly suppressing inflammatory damage to normal tissue. Therefore, opaganib may suppress toxicity from unintended IR exposure and improve patient response to chemoradiation. To test these hypotheses, we first examined the effects of opaganib on the toxicity and antitumor activity of radiation in mice exposed to total body irradiation (TBI) or IR with partial bone marrow shielding. Oral treatment with opaganib 2 h before TBI shifted the LD75 from 9.5 Gy to 11.5 Gy, and provided substantial protection against gastrointestinal damage associated with suppression of radiation-induced elevations of S1P and TNFα in the small intestines. In the partially shielded model, opaganib provided dose-dependent survival advantages when administered 4 h before or 24 h after radiation exposure, and was particularly effective when given both prior to and following radiation. Relevant to cancer radiotherapy, opaganib decreased the sensitivity of IEC6 (non-transformed mouse intestinal epithelial) cells to radiation, while sensitizing PAN02 cells to in vitro radiation. Next, the in vivo effects of opaganib in combination with radiation were examined in a syngeneic tumor model consisting of C57BL/6 mice bearing xenografts of PAN02 pancreatic cancer cells and a cross-species xenograft model consisting of nude mice bearing xenografts of human FaDu cells. Mice were treated with opaganib and/or IR (plus cisplatin in the case of FaDu tumors). In both tumor models, the optimal suppression of tumor growth was attained by the combination of opaganib with IR (± cisplatin). Overall, opaganib substantially protects normal tissue from radiation damage that may occur through unintended exposure or cancer radiotherapy.


Subject(s)
Cisplatin , Neoplasms , Humans , Mice , Animals , Mice, Nude , Mice, Inbred C57BL , Cell Line, Tumor
3.
Int J Nephrol Renovasc Dis ; 15: 323-334, 2022.
Article in English | MEDLINE | ID: mdl-36420520

ABSTRACT

Introduction: Acute kidney injury (AKI) is a common multifactorial adverse effect of surgery, circulatory obstruction, sepsis or drug/toxin exposure that often results in morbidity and mortality. Sphingolipid metabolism is a critical regulator of cell survival and pathologic inflammation processes involved in AKI. Opaganib (also known as ABC294640) is a first-in-class experimental drug targeting sphingolipid metabolism that reduces the production and activity of inflammatory cytokines and, therefore, may be effective to prevent and treat AKI. Methods: Murine models of AKI were used to assess the in vivo efficacy of opaganib including ischemia-reperfusion (IR) injury induced by either transient bilateral occlusion of renal blood flow (a moderate model) or nephrectomy followed immediately by occlusion of the contralateral kidney (a severe model) and lipopolysaccharide (LPS)-induced sepsis. Biochemical and histologic assays were used to quantify the effects of oral opaganib treatment on renal damage in these models. Results: Opaganib suppressed the elevations of creatinine and blood urea nitrogen (BUN), as well as granulocyte infiltration into the kidneys, of mice that experienced moderate IR from transient bilateral ligation. Opaganib also markedly decreased these parameters and completely prevented mortality in the severe renal IR model. Additionally, opaganib blunted the elevations of BUN, creatinine and inflammatory cytokines following exposure to LPS. Conclusion: The data support the hypotheses that sphingolipid metabolism is a key mediator of renal inflammatory damage following IR injury and sepsis, and that this can be suppressed by opaganib. Because opaganib has already undergone clinical testing in other diseases (cancer and Covid-19), the present studies support conducting clinical trials with this drug with surgical or septic patients at risk for AKI.

4.
J Pharmacol Exp Ther ; 365(1): 107-116, 2018 04.
Article in English | MEDLINE | ID: mdl-29434052

ABSTRACT

Glycogen synthase kinase-3s (GSK3α and GSK3ß) are constitutively active protein kinases that target over 100 substrates, incorporate into numerous protein complexes, and regulate such vital cellular functions as proliferation, apoptosis, and inflammation. Cyclin-dependent kinase 9 (CDK9) regulates RNA production as a component of positive transcription elongation factor b and promotes expression of oncogenic and inflammatory genes. Simultaneous inhibition of these signaling nodes is a promising approach for drug discovery, although previous compounds exhibit limited selectivity and clinical efficacy. The novel diaminothiazole ABC1183 is a selective GSK3α/ß and CDK9 inhibitor and is growth-inhibitory against a broad panel of cancer cell lines. ABC1183 treatment decreases cell survival through G2/M arrest and modulates oncogenic signaling through changes in GSK3, glycogen synthase, and ß-catenin phosphorylation and MCL1 expression. Oral administration, which demonstrates no organ or hematologic toxicity, suppresses tumor growth and inflammation-driven gastrointestinal disease symptoms, owing in part to downregulation of tumor necrosis factor α and interleukin-6 proinflammatory cytokines. Therefore, ABC1183 is strategically poised to effectively mitigate multiple clinically relevant diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Glycogen Synthase Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Humans , Inflammatory Bowel Diseases/drug therapy , Nitriles/therapeutic use , Protein Kinase Inhibitors/therapeutic use
5.
Pharmacology ; 87(3-4): 135-43, 2011.
Article in English | MEDLINE | ID: mdl-21346391

ABSTRACT

BACKGROUND/AIMS: Osteoarthritis (OA) is a progressive degenerative disease characterized by cartilage degradation and chondrocyte apoptosis, which may involve aberrant sphingolipid metabolism. ABC294640 is a compound that selectively inhibits sphingosine kinase-2, a key enzyme in the sphingolipid pathway. Our goal was to assess the pharmacological effects of ABC294640 in the monosodium iodoacetate (MIA) model of OA. METHODS: MIA (3 mg) was injected into the right knee joint to induce osteoarthritis in rats. Subsequently, the rats were treated with vehicle, ABC294640 or tramadol over a 28-day period. To assess pain, incapacitance readings were obtained weekly. MIA-injected knee joints were evaluated for histological damage, cartilage degradation and chondrocyte apoptosis (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling histochemistry). RESULTS: The percent weight bearing in vehicle/MIA rats significantly (p < 0.01) decreased from 48.8 ±0.8 (day 0) to 41.9 ±2.9 (day 28). In contrast, these values in ABC294640-treated rats were virtually the same on days 0 and 28. Knee joint histology scores were less severe in ABC294640-treated rats. Cartilage proteoglycan staining was more prominent in ABC294640/MIA animals than in vehicle/MIA rats. The percentage of apoptotic chondrocytes was decreased from 39.5% (vehicle treatment) to 25.8% (ABC294640 treatment). CONCLUSION: ABC294640 attenuated the knee joint histological damage and pain associated with MIA-induced OA in rats.


Subject(s)
Adamantane/analogs & derivatives , Osteoarthritis/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyridines/therapeutic use , Adamantane/administration & dosage , Adamantane/therapeutic use , Animals , Apoptosis/drug effects , Chondrocytes/drug effects , Chondrocytes/pathology , Disease Models, Animal , Iodoacetates/pharmacology , Male , Osteoarthritis/enzymology , Osteoarthritis/pathology , Pain/prevention & control , Pain Measurement , Pyridines/administration & dosage , Rats , Rats, Wistar
6.
Inflammopharmacology ; 19(2): 75-87, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20936538

ABSTRACT

UNLABELLED: Pro-inflammatory cytokines like TNF-α activate sphingosine kinase (SK). Therefore, inhibition of SK is a potential molecular target for the treatment of rheumatoid arthritis. AIMS: The primary goal of this study was to assess the efficacy of ABC249640 (a selective SK-2 inhibitor) in two models of rodent arthritis. A secondary goal was to evaluate the pharmacological profile of ABC294640, when given in combination with methotrexate. METHODS: The efficacy of ABC294640 was determined by paw diameter/volume measurements, histological evaluations, and micro-CT analyses. RESULTS: ABC294640 attenuated both collagen-induced arthritis in mice, as well as adjuvant-induced arthritis in rats. With the adjuvant arthritis model, the prophylactic efficacy profile of ABC294640 was similar to indomethacin. Of note, ABC294640 reduced the bone and cartilage degradation, associated with adjuvant-induced arthritis. Rats treated with a suboptimal dose of MTX (50 µg/kg/day) in combination with ABC249640 (100 mg/kg/day) had better anti-arthritis effects in the adjuvant model, than treatment with either agent alone. CONCLUSION: Our results suggest that ABC249640 is an orally available drug candidate with a good pre-clinical efficacy profile for the prevention and/or treatment of RA.


Subject(s)
Adamantane/analogs & derivatives , Arthritis, Experimental/drug therapy , Arthritis, Experimental/prevention & control , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyridines/administration & dosage , Pyridines/therapeutic use , Adamantane/administration & dosage , Adamantane/pharmacology , Adamantane/therapeutic use , Administration, Oral , Animals , Ankle/pathology , Ankle Joint/drug effects , Ankle Joint/pathology , Arthritis, Experimental/pathology , Body Weight/drug effects , Drug Therapy, Combination , Edema/pathology , Enzyme Inhibitors/pharmacology , Female , Foot/pathology , Indomethacin/administration & dosage , Indomethacin/pharmacology , Indomethacin/therapeutic use , Inflammation/pathology , Methotrexate/administration & dosage , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice , Mice, Inbred DBA , Pyridines/pharmacology , Rats , Rats, Inbred Lew , Splenomegaly/chemically induced , Splenomegaly/drug therapy , Splenomegaly/pathology , Splenomegaly/prevention & control , X-Ray Microtomography
7.
Inflammopharmacology ; 18(2): 73-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20151210

ABSTRACT

AIM: Activation of sphingosine kinase (SK) is a key response to many inflammatory processes. The present studies test the hypothesis that an orally available SK inhibitor, ABC294640, would be effective in rodent models of Crohn's disease. METHODS: Trinitrobenzene sulfonic acid (TNBS) was administered rectally to mice and rats. Rats were treated with ABC294640 orally alone or in combination with olsalazine and disease progression was monitored. RESULTS: For both rodent species, treatment with ABC294640 attenuated disease progression. Colon samples from the ABC294640-treated animals had improved histology and cytokine parameters when compared with vehicle-treated animals. The expression of SK was similarly increased in TNBS-treated animals and in human colon tissue specimens from inflammatory bowel disease patients relative to normal, control patients. CONCLUSIONS: Sphingosine kinase may be a critical mediator of colonic damage during intestinal inflammation, and pharmacologic inhibitors of this enzyme may prove useful in the treatment of Crohn's disease.


Subject(s)
Crohn Disease/chemically induced , Crohn Disease/drug therapy , Enzyme Inhibitors/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Adamantane/administration & dosage , Adamantane/analogs & derivatives , Adamantane/pharmacology , Adamantane/therapeutic use , Aminosalicylic Acids/administration & dosage , Aminosalicylic Acids/pharmacology , Aminosalicylic Acids/therapeutic use , Animals , Body Weight/drug effects , Colon/drug effects , Colon/enzymology , Colon/metabolism , Colon/pathology , Crohn Disease/metabolism , Crohn Disease/pathology , Disease Models, Animal , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/pharmacology , Gastrointestinal Agents/therapeutic use , Humans , Interleukin-1beta/metabolism , Leukocytes/enzymology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Neutrophils/enzymology , Neutrophils/pathology , Peroxidase/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prednisolone/administration & dosage , Prednisolone/pharmacology , Prednisolone/therapeutic use , Pyridines/administration & dosage , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Treatment Outcome , Trinitrobenzenesulfonic Acid/administration & dosage , Trinitrobenzenesulfonic Acid/pharmacology , Tumor Necrosis Factor-alpha/metabolism
8.
J Pharmacol Exp Ther ; 333(1): 129-39, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20061445

ABSTRACT

Sphingolipid-metabolizing enzymes control the dynamic balance of the cellular levels of important bioactive lipids, including the apoptotic compound ceramide and the proliferative compound sphingosine 1-phosphate (S1P). Many growth factors and inflammatory cytokines promote the cleavage of sphingomyelin and ceramide leading to rapid elevation of S1P levels through the action of sphingosine kinases (SK1 and SK2). SK1 and SK2 are overexpressed in a variety of human cancers, making these enzymes potential molecular targets for cancer therapy. We have identified an aryladamantane compound, termed ABC294640 [3-(4-chlorophenyl)-adamantane-1-carboxylic acid (pyridin-4-ylmethyl)amide], that selectively inhibits SK2 activity in vitro, acting as a competitive inhibitor with respect to sphingosine with a K(i) of 9.8 muM, and attenuates S1P formation in intact cells. In tissue culture, ABC294640 suppresses the proliferation of a broad panel of tumor cell lines, and inhibits tumor cell migration concomitant with loss of microfilaments. In vivo, ABC294640 has excellent oral bioavailability, and demonstrates a plasma clearance half-time of 4.5 h in mice. Acute and chronic toxicology studies indicate that ABC294640 induces a transient minor decrease in the hematocrit of rats and mice; however, this normalizes by 28 days of treatment. No other changes in hematology parameters, or gross or microscopic tissue pathology, result from treatment with ABC294640. Oral administration of ABC294640 to mice bearing mammary adenocarcinoma xenografts results in dose-dependent antitumor activity associated with depletion of S1P levels in the tumors and progressive tumor cell apoptosis. Therefore, this newly developed SK2 inhibitor provides an orally available drug candidate for the treatment of cancer and other diseases.


Subject(s)
Adamantane/analogs & derivatives , Antineoplastic Agents/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyridines/pharmacology , Adamantane/pharmacokinetics , Adamantane/pharmacology , Adamantane/therapeutic use , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Apoptosis , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotaxis/drug effects , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Transplantation , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
9.
J Surg Res ; 144(1): 1-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17640674

ABSTRACT

BACKGROUND: The claudins are tight junction (TJ) proteins. Claudin-2 has been found to negatively affect the TJ, causing a decrease in transepithelial resistance. Patients with inflammatory bowel disease have altered intestinal permeability, suggesting a TJ disruption. Interferon-gamma (IFNgamma) and interleukin-4 (IL-4) negatively regulate each other and may have opposing roles in inflammatory bowel disease. HYPOTHESIS: IFNgamma and IL-4 will have opposing effects on the expression of claudin-2. METHODS: Confluent T84 monolayers were apically incubated with IFNgamma or IL-4. The monolayers were immunofluorescently stained or lysed for Western blot with anti-claudin-2 or -4. Additional monolayers were grown on transwell plates, treated with IFNgamma or IL-4, measured for changes in transepithelial resistance, and assayed for changes in permeability using FITC-dextran-4000. Statistics were calculated by analysis of variance. RESULTS: Addition of IFNgamma to T84 monolayers resulted in decreased claudin-2 and addition of IL-4 resulted in increased claudin-2 by Western blot. By immunofluorescence, there was a loss of claudin-2 from the membrane in cells treated with IFNgamma. Transepithelial resistance across T84 monolayers increased with IFNgamma and decreased with IL-4. T84 monolayer permeability increased with IL-4 but not with IFNgamma. CONCLUSIONS: Incubation of T84 cells with IL-4 leads to increased claudin-2 with a corresponding decrease in transepithelial resistance and increase in permeability. Incubation of T84 cells with IFNgamma leads to decreased claudin-2 and increased transepithelial resistance. These cytokines have opposite effects on the expression of claudin-2 and the physiology of the TJ.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Membrane Proteins/metabolism , Tight Junctions/metabolism , Blotting, Western , Cell Line , Claudins , Electric Impedance , Fluorescent Antibody Technique , Humans , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Permeability/drug effects , Tight Junctions/drug effects , Tight Junctions/immunology
10.
J Surg Res ; 140(1): 12-9, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17418867

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is associated with increased intestinal permeability and decreased expression of tight junction (TJ) proteins in the inflamed mucosa. Whether this alteration in TJ expression is a prerequisite for the development of intestinal inflammation or a secondary result of that inflammation is unknown. This study looked at the expression of the TJ protein ZO-1 and the corresponding permeability changes in dextran sulfate sodium (DSS) induced colitis in a mouse model. MATERIALS AND METHODS: BALB/c mice were fed 3% DSS or water for 1, 3, 5, or 7 days. The animals were weighed, stool was checked for blood, and the colon length measured. Segments of the colon were used for histology, immunohistochemistry for ZO-1, or Western blot for TJ proteins. Colonic permeability was measured using Evan's Blue dye. RESULTS: DSS treated animals had heme positive stools, colitis by histology, significant weight loss, and colon shortening. There was an absence of ZO-1 by Western blot in the 7-day DSS treated animals, double the amount of claudin-1 and normal cytokeratin. The loss of ZO-1 started after 1 d of DSS treatment and was followed by a significant increase in permeability to Evan's blue by day 3. CONCLUSIONS: The loss of ZO-1 and increased permeability preceded the development of significant intestinal inflammation suggesting that in DSS colitis alterations in the TJ complex occur before the intestinal inflammation and not as a consequence of it. These changes in the TJ complex may facilitate the development of the inflammatory infiltrate seen in colitis.


Subject(s)
Colitis/metabolism , Colitis/pathology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Tight Junctions/metabolism , Tight Junctions/pathology , Animals , Anticoagulants , Blotting, Western , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Feces , Female , Fluorescent Antibody Technique , Gastrointestinal Hemorrhage/metabolism , Gastrointestinal Hemorrhage/pathology , Mice , Mice, Inbred BALB C , Weight Loss , Zonula Occludens-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...