1
|
Shen J, Hu C, Wang Y, Tan Y, Gao X, Zhang N, Lv J, Sun J. The SRC/NF-κB-AKT/NOS3 axis as a key mediator of Kaempferol's protective effects against oxidative stress-induced osteoclastogenesis. Immun Inflamm Dis 2024; 12:e70045. [PMID: 39422344 PMCID: PMC11488077 DOI: 10.1002/iid3.70045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Osteoclasts are integral to the advancement of osteoporosis (OP), and their generation under conditions of oxidative stress (OS) involves various pathways. However, the specific mechanism through which the natural antioxidant kaempferol (KAE) mitigates the influence of OS on osteoclasts remains somewhat uncertain. This study aims to evaluate the effect of KAE on osteoclast formation under OS and explore its possible mechanism. METHODS Zebrafish were used to observe the effects of KAE on OP and OS. OP and OS "double disease targets" network pharmacology were used to predict the action target and mechanism of KAE on OP under OS. The effects of KAE on osteoclast differentiation induced by OS were evaluated using RWA264.7 cells induced by LPS. To elucidate the potential mechanism, we detected the expression of related factors and target genes during induction. RESULTS The presence of KAE exhibited potential in improving the conditions of OP and OS in zebrafish. KAE can reduce the OS of RAW 264.7 cells stimulated by LPS, inhibit the formation of osteoclasts, and change the level of related factors of OS, and reduce the increase of TRAP. The utilization of network pharmacology and target gene expression assay revealed that KAE exerted a down-regulatory effect on the expression of proto-oncogene tyrosine protein kinase (SRC), nuclear factor kappa-B (NF-κB), Serine/Threonine Kinase-1 (AKT1), Nitric Oxide Synthase 3 (NOS3) and Matrix Metallopeptidase-2 (MMP2). CONCLUSION Based on the results of this study, KAE may effectively mitigate OS and impede the formation of osteoclasts through the SRC/NF-κB-AKT/NOS3 axis.
Collapse
Affiliation(s)
- Jiaming Shen
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Chunjie Hu
- Affiliated HospitalChangchun University of Chinese MedicineChangchunChina
| | - Yuelong Wang
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Yiying Tan
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Xiaochen Gao
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Nanxi Zhang
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Jingwei Lv
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| | - Jiaming Sun
- Jilin Ginseng AcademyChangchun University of Chinese MedicineChangchunChina
| |
Collapse
|
2
|
Lin X, Zhang Y, Huang H, Zhuang W, Wu L. Post-marketing safety concern of PI3K inhibitors in the cancer therapies: an 8-year disproportionality analysis from the FDA Adverse Event Reporting System. Expert Opin Drug Saf 2024:1-12. [PMID: 39083397 DOI: 10.1080/14740338.2024.2387317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/18/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The Phosphoinositide 3-kinases (PI3Ks) family plays a crucial role in tumorigenesis. Alpelisib (inhibiting PI3Kα), copanlisib (inhibiting PI3Kα andPI3Kδ), duvelisib (inhibiting PI3Kδ and PI3Kγ), and idelalisib (inhibiting PI3Kδ) were developed to target the PI3K pathway. However, the toxicity limits their application to some extent. It's necessary to investigate the adverse effects (AEs) of these inhibitors. RESEARCH DESIGN AND METHODS We conducted a comparative analysis of the safety signals of AEs in PI3K inhibitors using disproportionality analysis in the FDA Adverse Event Reporting System database(FAERS). RESULTS Our study identified significant safety signals for metabolic disorders with all PI3K inhibitors. Notable safety signals for gastrointestinal disorders were observed with most PI3K inhibitors, with the exception of copanlisib. Common AEs shared among all PI3K inhibitors included colitis and dehydration. Alpelisib displayed unique AEs associated with metabolic disorders, whereas copanlisib exhibited idiosyncratic AEs linked to cardiac and vascular disorders. Stevens-Johnson syndrome emerged as a common severe adverse event (SAE) among alpelisib, copanlisib, and idelalisib, while febrile neutropenia was prevalent among copanlisib, duvelisib, and idelalisib. Intestinal perforation was solely associated with alpelisib. CONCLUSIONS The safety profiles of the five PI3K inhibitors vary concerning adverse events. These findings could guide drug selection and inform future prospective research.
Collapse
Affiliation(s)
- Xiaorong Lin
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, Guangdong Province, China
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong Province, China
| | - Yimin Zhang
- Clinical Research Center, Shantou Central Hospital, Shantou, Guangdong Province, China
| | - Hongyan Huang
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wei Zhuang
- Department of Pharmacy, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian Province, China
| | - Lisha Wu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Department of Oncology, Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
| |
Collapse
|
3
|
Siddika T, Shao R, Heinemann IU, O'Donoghue P. Delivery of AKT1 phospho-forms to human cells reveals differential substrate selectivity. IUBMB Life 2024; 76:632-646. [PMID: 38738523 DOI: 10.1002/iub.2826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/25/2024] [Indexed: 05/14/2024]
Abstract
Protein kinase B (AKT1) is a serine/threonine kinase that regulates fundamental cellular processes, including cell survival, proliferation, and metabolism. AKT1 activity is controlled by two regulatory phosphorylation sites (Thr308, Ser473) that stimulate a downstream signaling cascade through phosphorylation of many target proteins. At either or both regulatory sites, hyperphosphorylation is associated with poor survival outcomes in many human cancers. Our previous biochemical and chemoproteomic studies showed that the phosphorylated forms of AKT1 have differential selectivity toward peptide substrates. Here, we investigated AKT1-dependent activity in human cells, using a cell-penetrating peptide (transactivator of transcription, TAT) to deliver inactive AKT1 or active phospho-variants to cells. We used enzyme engineering and genetic code expansion relying on a phosphoseryl-transfer RNA (tRNA) synthetase (SepRS) and tRNASep pair to produce TAT-tagged AKT1 with programmed phosphorylation at one or both key regulatory sites. We found that all TAT-tagged AKT1 variants were efficiently delivered into human embryonic kidney (HEK 293T) cells and that only the phosphorylated AKT1 (pAKT1) variants stimulated downstream signaling. All TAT-pAKT1 variants induced glycogen synthase kinase (GSK)-3α phosphorylation, as well as phosphorylation of ribosomal protein S6 at Ser240/244, demonstrating stimulation of downstream AKT1 signaling. Fascinatingly, only the AKT1 variants phosphorylated at S473 (TAT-pAKT1S473 or TAT-pAKT1T308,S473) were able to increase phospho-GSK-3β levels. Although each TAT-pAKT1 variant significantly stimulated cell proliferation, cells transduced with TAT-pAKT1T308 grew significantly faster than with the other pAKT1 variants. The data demonstrate differential activity of the AKT1 phospho-forms in modulating downstream signaling and proliferation in human cells.
Collapse
Affiliation(s)
- Tarana Siddika
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Richard Shao
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Ilka U Heinemann
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, The University of Western Ontario, London, Ontario, Canada
- Department of Chemistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
4
|
Rooban S, Arul Senghor K, Vinodhini V, Kumar J. Adropin: A crucial regulator of cardiovascular health and metabolic balance. Metabol Open 2024; 23:100299. [PMID: 39045137 PMCID: PMC11263719 DOI: 10.1016/j.metop.2024.100299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Adropin, a peptide discovered in 2008, has gained recognition as a key regulator of cardiovascular health and metabolic balance. Initially identified for its roles in energy balance, lipid metabolism, and glucose regulation, adropin has also been found to improve cardiovascular health by enhancing endothelial function, modulating lipid profiles, and reducing oxidative stress. These protective mechanisms suggest that adropin may be able to help prevent conditions such as atherosclerosis, hypertension, and other cardiovascular diseases. Research has established connections between adropin and cardiovascular risk factors, such as obesity, insulin resistance, and dyslipidemia, positioning it as a valuable biomarker for evaluating cardiovascular disease risk. New studies highlight adropin's diagnostic and prognostic significance, showing that higher levels are linked to better cardiovascular outcomes, while lower levels are associated with a higher risk of cardiovascular diseases. This review aims to summarize current knowledge on adropin, emphasizing its significance as a promising focus in the intersection of cardiovascular health and metabolic health. By summarizing the latest research findings, this review aims to offer insights into the potential applications of adropin in both clinical practice and research, leading to a deeper understanding of its role in maintaining cardiovascular and metabolic health.
Collapse
Affiliation(s)
- S. Rooban
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - K.A. Arul Senghor
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - V.M. Vinodhini
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| | - J.S. Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, 603203, Kanchipuram, Chennai, Tamil Nadu, India
| |
Collapse
|
5
|
Oost LJ, Tack CJ, de Baaij JHF. Hypomagnesemia and Cardiovascular Risk in Type 2 Diabetes. Endocr Rev 2022; 44:357-378. [PMID: 36346820 PMCID: PMC10166267 DOI: 10.1210/endrev/bnac028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/22/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022]
Abstract
Hypomagnesemia is tenfold more common in individuals with type 2 diabetes (T2D), compared to the healthy population. Factors that are involved in this high prevalence are low Mg2+ intake, gut microbiome composition, medication use and presumably genetics. Hypomagnesemia is associated with insulin resistance, which subsequently increases the risk to develop T2D or deteriorates glycaemic control in existing diabetes. Mg2+ supplementation decreases T2D associated features like dyslipidaemia and inflammation; which are important risk factors for cardiovascular disease (CVD). Epidemiological studies have shown an inverse association between serum Mg2+ and the risk to develop heart failure (HF), atrial fibrillation (AF) and microvascular disease in T2D. The potential protective effect of Mg2+ on HF and AF may be explained by reduced oxidative stress, fibrosis and electrical remodeling in the heart. In microvascular disease, Mg2+ reduces the detrimental effects of hyperglycemia and improves endothelial dysfunction. Though, clinical studies assessing the effect of long-term Mg2+ supplementation on CVD incidents are lacking and gaps remain on how Mg2+ may reduce CVD risk in T2D. Despite the high prevalence of hypomagnesemia in people with T2D, routine screening of Mg2+ deficiency to provide Mg2+ supplementation when needed is not implemented in clinical care as sufficient clinical evidence is lacking. In conclusion, hypomagnesemia is common in people with T2D and is both involved as cause, probably through molecular mechanisms leading to insulin resistance, and consequence and is prospectively associated with development of HF, AF and microvascular complications. Whether long-term supplementation of Mg2+ is beneficial, however, remains to be determined.
Collapse
Affiliation(s)
- Lynette J Oost
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
6
|
Fujie S, Hasegawa N, Horii N, Uchida M, Sanada K, Hamaoka T, Padilla J, Martinez-Lemus LA, Maeda S, Iemitsu M. Aerobic Exercise Restores Aging-Associated Reductions in Arterial Adropin Levels and Improves Adropin-Induced Nitric Oxide-Dependent Vasorelaxation. J Am Heart Assoc 2021; 10:e020641. [PMID: 33938228 PMCID: PMC8200711 DOI: 10.1161/jaha.120.020641] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Adropin is a peptide hormone that promotes nitric oxide (NO) production via activation of endothelial NO synthase (eNOS) in endothelial cells. Its circulating levels are reduced with aging and increased with aerobic exercise training (AT). Using a mouse model, we hypothesized that AT restores aging-associated reductions in arterial and circulating adropin and improves adropin-induced NO-dependent vasorelaxation. Further, we hypothesized these findings would be consistent with data obtained in elderly humans. Methods and Results In the animal study, 50-week-old SAMP1 male mice that underwent 12 weeks of voluntary wheel running, or kept sedentary, were studied. A separate cohort of 25-week-old SAMP1 male mice were used as a mature adult sedentary group. In the human study, 14 healthy elderly subjects completed an 8-week AT program consisting of 45 minutes of cycling 3 days/week. In mice, we show that advanced age is associated with a decline in arterial and circulating levels of adropin along with deterioration of endothelial function, arterial NO production, and adropin-induced vasodilation. All these defects were restored by AT. Moreover, AT-induced increases in arterial adropin were correlated with increases in arterial eNOS phosphorylation and NO production. Consistently with these findings in mice, AT in elderly subjects enhanced circulating adropin levels and these effects were correlated with increases in circulating nitrite/nitrate (NOx) and endothelial function. Conclusions Changes in arterial adropin that occur with age or AT relate to alterations in endothelial function and NO production, supporting the notion that adropin should be considered a therapeutic target for vascular aging. Registration URL: https://www.umin.ac.jp; Unique identifier: UMIN000035520.
Collapse
Affiliation(s)
- Shumpei Fujie
- Faculty of Sport and Health Science Ritsumeikan University Shiga Japan.,Faculty of Sport and Health Sciences University of Tsukuba Ibaraki Japan.,Dalton Cardiovascular Research Center University of Missouri MO
| | - Natsuki Hasegawa
- Research Organization of Science and Technology Ritsumeikan University Shiga Japan
| | - Naoki Horii
- Faculty of Sport and Health Science Ritsumeikan University Shiga Japan.,Research Fellow of Japan Society for the Promotion of Science Tokyo Japan
| | - Masataka Uchida
- Faculty of Sport and Health Science Ritsumeikan University Shiga Japan
| | - Kiyoshi Sanada
- Faculty of Sport and Health Science Ritsumeikan University Shiga Japan
| | - Takafumi Hamaoka
- Sports Medicine for Health Promotion Tokyo Medical University Tokyo Japan
| | - Jaume Padilla
- Dalton Cardiovascular Research Center University of Missouri MO.,Nutrition and Exercise Physiology University of Missouri MO
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center University of Missouri MO.,Medical Pharmacology and Physiology University of Missouri MO
| | - Seiji Maeda
- Faculty of Sport and Health Sciences University of Tsukuba Ibaraki Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science Ritsumeikan University Shiga Japan
| |
Collapse
|
7
|
Munoz J, Follows GA, Nastoupil LJ. Copanlisib for the Treatment of Malignant Lymphoma: Clinical Experience and Future Perspectives. Target Oncol 2021; 16:295-308. [PMID: 33687623 PMCID: PMC7941125 DOI: 10.1007/s11523-021-00802-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2021] [Indexed: 01/17/2023]
Abstract
Dysregulation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B/mammalian target of rapamycin signaling is common in both indolent and aggressive forms of malignant lymphoma, for which several targeted therapies have been developed. Copanlisib is a highly selective and potent intravenous pan-class I PI3K inhibitor that has demonstrated durable objective responses and a manageable safety profile in heavily pre-treated patients with indolent lymphomas. As a result, copanlisib monotherapy received accelerated approval from the US Food and Drug Administration for the treatment of adults with relapsed follicular lymphoma who have received at least two systemic therapies, and breakthrough designation for patients with pre-treated relapsed or refractory marginal zone lymphoma. Hyperglycemia and hypertension are among the most frequently reported adverse events with copanlisib monotherapy, and are infusion-related, transient, and manageable with standard therapies. Mild diarrhea is also a common adverse event with copanlisib monotherapy; there is no evidence of worsening severity of diarrhea, or serious gastrointestinal toxicities such as colitis or severe liver enzyme elevations, which have been reported with orally administered PI3K inhibitors. The intravenous route of administration and intermittent dosing schedule of copanlisib may support a favorable tolerability profile over continually administered oral alternatives. Ongoing studies of copanlisib in combination with rituximab and standard-of-care chemotherapy in patients with relapsed indolent lymphoma have the potential to support the use of copanlisib in the second-line setting, providing a much-needed additional therapeutic option in this underserved patient population.
Collapse
Affiliation(s)
- Javier Munoz
- Mayo Clinic, 5881 E. Mayo Boulevard, Phoenix, AZ, 85054, USA.
| | - George A Follows
- Department of Haematology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Loretta J Nastoupil
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Morschhauser F, Machiels JP, Salles G, Rottey S, Rule SAJ, Cunningham D, Peyrade F, Fruchart C, Arkenau HT, Genvresse I, Liu L, Köchert K, Shen K, Kneip C, Peña CE, Grevel J, Zhang J, Cisternas G, Reschke S, Granvil C, Awada A. On-Target Pharmacodynamic Activity of the PI3K Inhibitor Copanlisib in Paired Biopsies from Patients with Malignant Lymphoma and Advanced Solid Tumors. Mol Cancer Ther 2019; 19:468-478. [PMID: 31619463 DOI: 10.1158/1535-7163.mct-19-0466] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/12/2019] [Accepted: 10/07/2019] [Indexed: 11/16/2022]
Abstract
The PI3K inhibitor copanlisib has efficacy and manageable safety in patients with indolent lymphoma and solid tumors. Pharmacodynamic effects relative to copanlisib dose and plasma exposure were evaluated. Patients with lymphoma or solid tumors received copanlisib 0.4 or 0.8 mg/kg on days 1, 8, and 15 of a 28-day cycle. Primary variables were maximum changes in phosphorylated AKT (pAKT) levels in platelet-rich plasma (PRP) and plasma glucose. Other evaluations included PI3K signaling markers and T-lymphocytes in paired tumor biopsies, the relationship between estimated plasma exposure and pharmacodynamic markers, response, and safety. Sixty-three patients received copanlisib. PRP pAKT levels showed sustained reductions from baseline following copanlisib [median inhibition: 0.4 mg/kg, 73.8% (range -94.9 to 144.0); 0.8 mg/kg, 79.6% (range -96.0 to 408.0)]. Tumor pAKT was reduced versus baseline with copanlisib 0.8 mg/kg in paired biopsy samples (P < 0.05). Dose-related transient plasma glucose elevations were observed. Estimated copanlisib plasma exposure significantly correlated with changes in plasma pAKT and glucose metabolism markers. There were two complete responses and six partial responses; seven of eight responders received copanlisib 0.8 mg/kg. Adverse events (all grade) included hyperglycemia (52.4%), fatigue (46.0%), and hypertension (41.3%). Copanlisib demonstrated dose-dependent pharmacodynamic evidence of target engagement and PI3K pathway modulation/inhibition in tumor and immune cells. Results support the use of copanlisib 0.8 mg/kg (or flat-dose equivalent of 60 mg) in solid tumors and lymphoma, and provide a biomarker hypothesis for studies of copanlisib combined with immune checkpoint inhibitors (NCT03711058).
Collapse
Affiliation(s)
- Franck Morschhauser
- Service des Maladies du Sang, Université de Lille, CHU Lille, Lille, France.
| | - Jean-Pascal Machiels
- Service d'Oncologie Médicale, Institut Roi Albert II, Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale, UCLouvain, Brussels, Belgium
| | - Gilles Salles
- Service d'Hématologie, Clinique Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
- Hospices Civils de Lyon, Lyon, France
| | - Sylvie Rottey
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Simon A J Rule
- Department of Haematology, Plymouth University Medical School, Plymouth Hospitals NHS Trust, Plymouth, United Kingdom
| | - David Cunningham
- Department of Clinical and Experimental Haematology, The Royal Marsden Hospital, Sutton, United Kingdom
| | - Frederic Peyrade
- Department of Medical Oncology, Centre Antoine Lacassagne, Nice, France
| | - Christophe Fruchart
- Institut d'Hématologie de Basse Normandie, Centre Hospitalier Universitaire, Caen, France
| | | | | | - Li Liu
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | - Karl Köchert
- Pharmaceuticals Division, Bayer AG, Berlin, Germany
| | - Kui Shen
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | - Carol E Peña
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | - Jun Zhang
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | | | - Camille Granvil
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | - Ahmad Awada
- Clinique Oncologie Médicale, Institut Jules Bordet, Brussels, Belgium
| |
Collapse
|
9
|
van Roeyen CRC, Martin IV, Drescher A, Schuett KA, Hermert D, Raffetseder U, Otten S, Buhl EM, Braun GS, Kuppe C, Liehn E, Boor P, Weiskirchen R, Eriksson U, Gross O, Eitner F, Floege J, Ostendorf T. Identification of platelet-derived growth factor C as a mediator of both renal fibrosis and hypertension. Kidney Int 2019; 95:1103-1119. [PMID: 30827511 DOI: 10.1016/j.kint.2018.11.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 11/15/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Platelet-derived growth factors (PDGF) have been implicated in kidney disease progression. We previously found that PDGF-C is upregulated at sites of renal fibrosis and that antagonism of PDGF-C reduces fibrosis in the unilateral ureteral obstruction model. We studied the role of PDGF-C in collagen 4A3-/- ("Alport") mice, a model of progressive renal fibrosis with greater relevance to human kidney disease. Alport mice were crossbred with PDGF-C-/- mice or administered a neutralizing PDGF-C antibody. Both PDGF-C deficiency and neutralization reduced serum creatinine and blood urea nitrogen levels and mitigated glomerular injury, renal fibrosis, and renal inflammation. Unexpectedly, systolic blood pressure was also reduced in both Alport and wild-type mice treated with a neutralizing PDGF-C antibody. Neutralization of PDGF-C reduced arterial wall thickness in the renal cortex of Alport mice. Aortic rings isolated from anti-PDGF-C-treated wildtype mice exhibited reduced tension and faster relaxation than those of untreated mice. In vitro, PDGF-C upregulated angiotensinogen in aortic tissue and in primary hepatocytes and induced nuclear factor κB (NFκB)/p65-binding to the angiotensinogen promoter in hepatocytes. Neutralization of PDGF-C suppressed transcript expression of angiotensinogen in Alport mice and angiotensin II receptor type 1 in Alport and wildtype mice. Finally, administration of neutralizing PDGF-C antibodies ameliorated angiotensin II-induced hypertension in healthy mice. Thus, in addition to its key role in mediating renal fibrosis, we identified PDGF-C as a mediator of hypertension via effects on renal vasculature and on the renin-angiotensin system. The contribution to both renal fibrosis and hypertension render PDGF-C an attractive target in progressive kidney disease.
Collapse
Affiliation(s)
- Claudia R C van Roeyen
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| | - Ina V Martin
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ana Drescher
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | | | - Daniela Hermert
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ute Raffetseder
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Stephanie Otten
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Eva M Buhl
- Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Gerald S Braun
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Elisa Liehn
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Peter Boor
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry RWTH Aachen University, Aachen, Germany
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Oliver Gross
- Division of Nephrology and Rheumatology, University Medicine Göttingen, Göttingen, Germany
| | - Frank Eitner
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany; Kidney Diseases Research, Bayer AG, Wuppertal, Germany
| | - Jürgen Floege
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Tammo Ostendorf
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
10
|
Patnaik A, Appleman LJ, Tolcher AW, Papadopoulos KP, Beeram M, Rasco DW, Weiss GJ, Sachdev JC, Chadha M, Fulk M, Ejadi S, Mountz JM, Lotze MT, Toledo FGS, Chu E, Jeffers M, Peña C, Xia C, Reif S, Genvresse I, Ramanathan RK. First-in-human phase I study of copanlisib (BAY 80-6946), an intravenous pan-class I phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors and non-Hodgkin's lymphomas. Ann Oncol 2017; 27:1928-40. [PMID: 27672108 PMCID: PMC5035790 DOI: 10.1093/annonc/mdw282] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND To evaluate the safety, tolerability, pharmacokinetics, and maximum tolerated dose (MTD) of copanlisib, a phosphatidylinositol 3-kinase inhibitor, in patients with advanced solid tumors or non-Hodgkin's lymphoma (NHL). PATIENTS AND METHODS Phase I dose-escalation study including patients with advanced solid tumors or NHL, and a cohort of patients with type 2 diabetes mellitus. Patients received three weekly intravenous infusions of copanlisib per 28-day cycle over the dose range 0.1-1.2 mg/kg. Plasma copanlisib levels were analyzed for pharmacokinetics. Biomarker analysis included PIK3CA, KRAS, BRAF, and PTEN mutational status and PTEN immunohistochemistry. Whole-body [(18)F]-fluorodeoxyglucose positron emission tomography ((18)FDG-PET) was carried out at baseline and following the first dose to assess early pharmacodynamic effects. Plasma glucose and insulin levels were evaluated serially. RESULTS Fifty-seven patients received treatment. The MTD was 0.8 mg/kg copanlisib. The most frequent treatment-related adverse events were nausea and transient hyperglycemia. Copanlisib exposure was dose-proportional with no accumulation; peak exposure positively correlated with transient hyperglycemia post-infusion. Sixteen of 20 patients treated at the MTD had reduced (18)FDG-PET uptake; 7 (33%) had a reduction >25%. One patient achieved a complete response (CR; endometrial carcinoma exhibiting both PIK3CA and PTEN mutations and complete PTEN loss) and two had a partial response (PR; both metastatic breast cancer). Among the nine NHL patients, all six with follicular lymphoma (FL) responded (one CR and five PRs) and one patient with diffuse large B-cell lymphoma had a PR by investigator assessment; two patients with FL who achieved CR (per post hoc independent radiologic review) were on treatment >3 years. CONCLUSION Copanlisib, dosed intermittently on days 1, 8, and 15 of a 28-day cycle, was well tolerated and the MTD was determined to be 0.8 mg/kg. Copanlisib exhibited dose-proportional pharmacokinetics and promising anti-tumor activity, particularly in patients with NHL. CLINICALTRIALSGOV NCT00962611; https://clinicaltrials.gov/ct2/show/NCT00962611.
Collapse
Affiliation(s)
- A Patnaik
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | | | - A W Tolcher
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - K P Papadopoulos
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - M Beeram
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - D W Rasco
- South Texas Accelerated Research Therapeutics (START) Center for Cancer Care, San Antonio
| | - G J Weiss
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale Cancer Treatment Centers of America, Goodyear
| | - J C Sachdev
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - M Chadha
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - M Fulk
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | - S Ejadi
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| | | | - M T Lotze
- University of Pittsburgh, Pittsburgh
| | | | - E Chu
- University of Pittsburgh, Pittsburgh
| | - M Jeffers
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - C Peña
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - C Xia
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, USA
| | - S Reif
- Bayer Pharma AG, Berlin, Germany
| | | | - R K Ramanathan
- Virginia G. Piper Cancer Center Clinical Trials at Scottsdale Healthcare/TGen, Scottsdale
| |
Collapse
|
11
|
Murthy S, Koval OM, Ramiro Diaz JM, Kumar S, Nuno D, Scott JA, Allamargot C, Zhu LJ, Broadhurst K, Santhana V, Kutschke WJ, Irani K, Lamping KG, Grumbach IM. Endothelial CaMKII as a regulator of eNOS activity and NO-mediated vasoreactivity. PLoS One 2017; 12:e0186311. [PMID: 29059213 PMCID: PMC5653296 DOI: 10.1371/journal.pone.0186311] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 09/28/2017] [Indexed: 01/11/2023] Open
Abstract
The multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a serine/threonine kinase important in transducing intracellular Ca2+ signals. While in vitro data regarding the role of CaMKII in the regulation of endothelial nitric oxide synthase (eNOS) are contradictory, its role in endothelial function in vivo remains unknown. Using two novel transgenic models to express CaMKII inhibitor peptides selectively in endothelium, we examined the effect of CaMKII on eNOS activation, NO production, vasomotor tone and blood pressure. Under baseline conditions, CaMKII activation was low in the aortic wall. Consistently, systolic and diastolic blood pressure, heart rate and plasma NO levels were unaltered by endothelial CaMKII inhibition. Moreover, endothelial CaMKII inhibition had no significant effect on NO-dependent vasodilation. These results were confirmed in studies of aortic rings transduced with adenovirus expressing a CaMKII inhibitor peptide. In cultured endothelial cells, bradykinin treatment produced the anticipated rapid influx of Ca2+ and transient CaMKII and eNOS activation, whereas CaMKII inhibition blocked eNOS phosphorylation on Ser-1179 and dephosphorylation at Thr-497. Ca2+/CaM binding to eNOS and resultant NO production in vitro were decreased under CaMKII inhibition. Our results demonstrate that CaMKII plays an important role in transient bradykinin-driven eNOS activation in vitro, but does not regulate NO production, vasorelaxation or blood pressure in vivo under baseline conditions.
Collapse
Affiliation(s)
- Shubha Murthy
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
| | - Olha M. Koval
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
| | - Juan M. Ramiro Diaz
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Santosh Kumar
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Daniel Nuno
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Jason A. Scott
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Chantal Allamargot
- Central Microscopy Research Facility, Office of Vice President of Research and Economic Development, University of Iowa, Iowa City, Iowa, United States of America
| | - Linda J. Zhu
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Kim Broadhurst
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Velarchana Santhana
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - William J. Kutschke
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Kaikobad Irani
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
| | - Kathryn G. Lamping
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Isabella M. Grumbach
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Iowa City Veterans Affairs Healthcare System, Iowa City, Iowa, United States of America
| |
Collapse
|
12
|
Arcucci A, Ruocco MR, Albano F, Granato G, Romano V, Corso G, Bancone C, De Vendittis E, Della Corte A, Montagnani S. Analysis of extracellular superoxide dismutase and Akt in ascending aortic aneurysm with tricuspid or bicuspid aortic valve. Eur J Histochem 2014; 58:2383. [PMID: 25308842 PMCID: PMC4194390 DOI: 10.4081/ejh.2014.2383] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 11/23/2022] Open
Abstract
Ascending aortic aneurysm (AsAA) is a consequence of medial degeneration (MD), deriving from apoptotic loss of smooth muscle cells (SMC) and fragmentation of elastin and collagen fibers. Alterations of extracellular matrix structure and protein composition, typical of medial degeneration, can modulate intracellular pathways. In this study we examined the relevance of superoxide dismutase (SOD3) and Akt in AsAA pathogenesis, evaluating their tissue distribution and protein levels in ascending aortic tissues from controls (n=6), patients affected by AsAA associated to tricuspid aortic valve (TAV, n=9) or bicuspid aortic valve (BAV, n=9). The results showed a significant reduction of SOD3, phospho-Akt and Akt protein levels in AsAA tissues from patients with BAV, compared to controls, whereas the differences observed between controls and patients with TAV were not significant. The decreased levels of SOD3 and Akt in BAV aortic tissues are associated with decreased Erk1/Erk2 phosphorylation and MMP-9 levels increase. The authors suggest a role of decreased SOD3 protein levels in the progression of AsAA with BAV and a link between ECM modifications of aortic media layer and impaired Erk1/Erk2 and Akt signaling in the late stages of the aortopathy associated with BAV.
Collapse
|
13
|
Jin SY, Kim EK, Ha JM, Lee DH, Kim JS, Kim IY, Song SH, Shin HK, Kim CD, Bae SS. Insulin regulates monocyte trans-endothelial migration through surface expression of macrophage-1 antigen. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1539-48. [PMID: 24915517 DOI: 10.1016/j.bbadis.2014.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 11/16/2022]
Abstract
During the pathogenesis of atherosclerosis, adhesion of monocytes to vascular endothelium and subsequent migration across the endothelium has been recognized as a key process in the chronic inflammatory response in atherosclerosis. As type 2 diabetes is closely associated with the pathogenesis of atherosclerosis, we investigated whether monocyte adhesion and migration were affected by insulin. We found that insulin activated Akt and induced subsequent migration in THP-1. However, glucose and insulin-like growth factor-1, which is a growth factor that is structurally similar to insulin, were not effective. Insulin-dependent migration of THP-1 was blocked by inhibition of PI3K or Akt and by silencing of Akt1. Insulin-dependent migration of bone marrow-derived monocytic cells (BDMCs) was attenuated by inhibition of PI3K and Akt. In addition, BDMCs from Akt1(-/-) mice showed defects in insulin-dependent migration. Stimulation of THP-1 with insulin caused adhesion with human vein endothelial cells (HUVECs) that was blocked by silencing of Akt1. However, stimulation of HUVECs did not cause adhesion with THP-1. Moreover, BDMCs from Akt1(-/-) mice showed defects in insulin-dependent adhesion with HUVECs. Insulin induced surface expression of Mac-1, and neutralization of Mac-1 blocked insulin-induced adhesion of THP-1 as well as BDMCs. Surface expression of Mac-1 was blocked in THP-1 with silenced Akt1, and in BDMCs isolated from mice lacking Akt1. Finally, trans-endothelial migration of THP-1 and BDMCs was blocked by Mac-1-neutralizing antibody, in THP-1 with silenced Akt1 and in BDMCs from Akt1(-/-) mice. These results suggest that insulin stimulates monocyte trans-endothelial migration through Akt-dependent surface expression of Mac-1, which may be part of the atherogenesis in type 2 diabetes.
Collapse
Affiliation(s)
- Seo Yeon Jin
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Eun Kyoung Kim
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jung Min Ha
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Hyung Lee
- Department of Obstetrics and Gynecology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jeong Su Kim
- Cardiovascular Disease Center, Pusan National University Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Il Young Kim
- Department of Internal Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sang Heon Song
- Department of Internal Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Anatomy, Pusan National University School of Korean Medicine, Yangsan, Republic of Korea
| | - Chi Dae Kim
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sun Sik Bae
- MRC for Ischemic Tissue Regeneration, Medical Research Institute, and Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Republic of Korea.
| |
Collapse
|