Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacol Res ; 107: 372-380, 2016 05.
Article in English | MEDLINE | ID: mdl-26956523

ABSTRACT

Congestive heart failure is one of the most prevalent and deadly complications of type 2 diabetes that is frequently associated with pulmonary dysfunction. Among many factors that contribute to development and progression of diabetic complications is angiotensin II (Ang2). Activation of pathological arm of renin-angiotensin system results in increased levels of Ang2 and signaling through angiotensin type 1 receptor. This pathway is well recognized for its role in induction of oxidative stress (OS), inflammation, hypertrophy and fibrosis. Angiotensin (1-7) [A(1-7)], through activation of Mas receptor, opposes the actions of Ang2 which can result in the amelioration of diabetic complications; enhancing the overall welfare of diabetic patients. In this study, 8 week-old db/db mice were administered A(1-7) daily via subcutaneous injections. After 16 weeks of treatment, echocardiographic assessment of heart function demonstrated significant improvement in cardiac output, stroke volume and shortening fraction in diabetic animals. A(1-7) also prevented cardiomyocyte hypertrophy, apoptosis, lipid accumulation, and decreased diabetes-induced fibrosis and OS in the heart tissue. Treatment with A(1-7) reduced levels of circulating proinflammatory cytokines that contribute to the low grade inflammation observed in diabetes. In addition, lung pathologies associated with type 2 diabetes, including fibrosis and congestion, were decreased with treatment. OS and macrophage infiltration were also reduced in the lungs after treatment with A(1-7). Long-term administration of A(1-7) to db/db mice is effective in improving heart and lung function in db/db mice. Treatment prevented pathological remodeling of the tissues and reduced OS, fibrosis and inflammation.


Subject(s)
Angiotensin I/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Peptide Fragments/therapeutic use , Angiotensin I/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Cytokines/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Fibrosis , Heart/drug effects , Heart/physiology , Hypoglycemic Agents/pharmacology , Lipid Metabolism/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/drug effects , Peptide Fragments/pharmacology
2.
Molecules ; 21(2): 152, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26821010

ABSTRACT

We report for the first time the design and synthesis of a novel cyclotide able to activate the unique receptor of angiotensin (1-7) (AT1-7), the MAS1 receptor. This was accomplished by grafting an AT1-7 peptide analog onto loop 6 of cyclotide MCoTI-I using isopeptide bonds to preserve the α-amino and C-terminal carboxylate groups of AT1-7, which are required for activity. The resulting cyclotide construct was able to adopt a cyclotide-like conformation and showed similar activity to that of AT1-7. This cyclotide also showed high stability in human serum thereby providing a promising lead compound for the design of a novel type of peptide-based in the treatment of cancer and myocardial infarction.


Subject(s)
Cyclotides/chemical synthesis , Cyclotides/pharmacology , Plant Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Angiotensin I/chemistry , Angiotensin I/pharmacology , Animals , CHO Cells , Cell Survival/drug effects , Cricetulus , Cyclotides/chemistry , Humans , Myocardial Infarction/drug therapy , Neoplasms/drug therapy , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Protein Conformation , Protein Folding , Protein Stability , Proto-Oncogene Mas
3.
Apoptosis ; 20(7): 892-906, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25772546

ABSTRACT

In the event of a nuclear disaster, the individuals proximal to the source of radiation will be exposed to combined radiation injury. As irradiation delays cutaneous repair, the purpose of this study was to elucidate the effect of combined radiation and burn injury (CRBI) on apoptosis and inflammation at the site of skin injury. Male C57Bl/6 mice were exposed to no injury, thermal injury only, radiation only (1 and 6 Gy) and CRBI (1 and 6 Gy) and euthanized at various times after for skin collection. TUNEL staining revealed that the CRBI 6 Gy group had a delayed and increased apoptotic response. This correlated with decreased recovery of live cells as compared to the other injuries. Similar response was observed when cleaved-caspase-3 immunohistochemical staining was compared between CRBI 6 Gy and thermal injury. TNFR1, caspase 8, Bax and IL-6 mRNA expression revealed that the higher CRBI group had delayed increase in mRNA expression as compared to thermal injury alone. RIPK1 mRNA expression and necrotic cell counts were delayed in the CRBI 6 Gy group to day 5. TNF-α and NFκB expression peaked in the CRBI 6 Gy group at day 1 and was much higher than the other injuries. Also, inflammatory cell counts in the CRBI 6 Gy group were lower at early time points as compared to thermal injury by itself. These data suggest that CRBI delays and exacerbates apoptosis and inflammation in skin as well as increases necrosis thus resulting in delayed wound healing.


Subject(s)
Apoptosis/radiation effects , Burns/pathology , Radiation Injuries, Experimental/pathology , Radiation Injuries/metabolism , Skin/radiation effects , Animals , Biomarkers/metabolism , Burns/metabolism , Inflammation/metabolism , Male , Mice, Inbred C57BL , Necrosis/metabolism , Radiation Injuries, Experimental/metabolism , Skin/metabolism , Transcriptome
4.
Cancer Chemother Pharmacol ; 72(6): 1235-45, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24091850

ABSTRACT

PURPOSE: Angiotensin (1-7) [A(1-7)] is a bioactive peptide of the renin angiotensin system that stimulates the number of bone marrow progenitors and hematopoietic recovery after myelosuppression. We evaluated the combination of A(1-7) with colony-stimulating factors, Neupogen and Epogen, on bone marrow progenitors and the recovery of circulating formed elements following chemotherapy. METHODS: Mice were injected with gemcitabine followed 2 days later with A(1-7). Circulating blood cells and bone marrow progenitors were measured over time. RESULTS: Combination of A(1-7) with Neupogen (the latter given only 3 days starting at the white blood cell nadir) decreased the amount of Neupogen needed for optimal recovery by 10-fold. The progenitors measured include CFU-GEMM, CFU-GM, CFU-Meg and BFU-E. A(1-7) increased recovery of all progenitors when given alone or in combination with Neupogen above that with Neupogen alone. Combination of A(1-7) with Epogen slightly increased (not significantly) red blood cell concentrations above those achieved by Epogen alone. However, in this model, A(1-7) or A(1-7) in combination with Epogen increased all erythroid progenitors with the largest effect on early erythroid progenitors (immature BFU-E). CONCLUSIONS: Neupogen and Epogen acted synergistically with A(1-7) to increase the concentration of myeloid, megakaryocytic and erythroid progenitor cells in the bone marrow following chemotherapy suggesting that A(1-7)'s multilineage effect on early progenitors in the marrow facilitates proliferation in response to lineage-specific growth factors.


Subject(s)
Angiotensin I/pharmacology , Antimetabolites, Antineoplastic/toxicity , Deoxycytidine/analogs & derivatives , Erythropoietin/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacology , Peptide Fragments/pharmacology , Angiotensin I/administration & dosage , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Deoxycytidine/toxicity , Drug Synergism , Epoetin Alfa , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/administration & dosage , Female , Filgrastim , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematinics , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Peptide Fragments/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Time Factors , Gemcitabine
5.
Wound Repair Regen ; 21(1): 131-40, 2013.
Article in English | MEDLINE | ID: mdl-23231670

ABSTRACT

The renin-angiotensin system (RAS) plays an important role in wound repair; however, little is known pertaining to RAS expression in response to thermal injury and the combination of radiation plus burn injury (CRBI). The purpose of this study was to test the hypothesis that thermal injury modifies expression of RAS components and CRBI delayed this up-regulation of RAS. Skin from uninjured mice was compared with mice receiving local thermal injury or CRBI (injury site). Skin was analyzed for gene and protein expression of RAS components. There was an initial increase in the expression of various components of RAS following thermal injury. However, in the higher CRBI group there is an initial decrease in AT(1b) (vasoconstriction, pro-proliferative), AT(2) (vasodilation, differentiation), and Mas (vasodilation, anti-inflammatory) gene expression. This corresponded with a delay and decrease in AT(1) , AT(2) , and MAS protein expression in fibroblasts and keratinocytes. The reduction in RAS receptor positive fibroblasts and keratinocytes correlated with a reduction in collagen deposition and keratinocyte infiltration into the wounded area resulting in a delay of reepithelialization following CRBI. These data support the hypothesis that delayed wound healing observed in subjects following radiation exposure may be in part due to decreased expression of RAS.


Subject(s)
Burns/metabolism , Collagen/metabolism , Radiation Injuries, Experimental/metabolism , Renin-Angiotensin System , Skin/metabolism , Wound Healing , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II Type 2 Receptor Blockers/metabolism , Animals , Burns/pathology , Disease Models, Animal , Female , Fibroblasts/metabolism , Gene Expression , Immunohistochemistry , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology , Skin/injuries , Skin/pathology , Up-Regulation
6.
Int J Radiat Biol ; 88(6): 466-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22433112

ABSTRACT

PURPOSE: Angiotensin (1-7) [A(1-7)] is a component of the renin angiotensin system (RAS) that stimulates hematopoietic recovery after myelosuppression. In a Phase I/IIa clinical trial, thrombocytopenia after chemotherapy was reduced by A(1-7). In this study, the ability of A(1-7) to improve recovery after total body irradiation (TBI) is shown with specific attention to radiation-induced hematopoietic injury. MATERIALS AND METHODS: Mice were exposed to TBI (doses of 2-7 Gray [Gy]) of cesium 137 gamma rays, followed by treatment with A(1-7), typical doses were 100-1000 µg/kg given once or once daily for a specified number of days depending on the study. Animals are injected subcutaneously via the nape of the neck with 0.1 ml drug in saline. The recovery of blood and bone marrow cells was determined. Effects of TBI and A(1-7) on survival and bleeding time was also evaluated. RESULTS: Daily administration of A(1-7) after radiation exposure improved survival (from 60% to 92-97%) and reduced bleeding time at day 30 after TBI. Further, A(1-7) increased early mixed progenitors (3- to 5-fold), megakaryocyte (2- to 3-fold), myeloid (3- to 6-fold) and erythroid (2- to 5-fold) progenitors in the bone marrow and reduced radiation-induced thrombocytopenia (RIT) (up to 2-fold). Reduction in the number of treatments to 3 per week also improved bone marrow recovery and reduced RIT. As emergency responder and healthcare systems in case of nuclear accident or/and terrorist attack may be overwhelmed, the consequence of delayed initiation of treatment was ascertained. Treatment with A(1-7) can be delayed up to 5 days and still be effective in the reduction of RIT or acceleration of bone marrow recovery. CONCLUSIONS: The data presented in this paper indicate that A(1-7) reduces the consequences of critical radiation exposure and can be initiated well after initial exposure with maximal effects on early responding hematopoietic progenitors when treatment is initiated 2 days after exposure and 5 days after exposure for the later responding progenitors and reduced thrombocytopenia. There was some effect of A(1-7) even when given days after radiation exposure.


Subject(s)
Angiotensin I/pharmacology , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Peptide Fragments/pharmacology , Whole-Body Irradiation , Anemia/etiology , Anemia/prevention & control , Angiotensin I/administration & dosage , Animals , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Administration Schedule , Female , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Leukopenia/etiology , Leukopenia/prevention & control , Mice , Mice, Inbred C57BL , Peptide Fragments/administration & dosage , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/pharmacology , Renin-Angiotensin System/physiology , Thrombocytopenia/etiology , Thrombocytopenia/prevention & control , Whole-Body Irradiation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...