Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Publication year range
1.
J Cell Physiol ; 239(5): e31212, 2024 May.
Article in English | MEDLINE | ID: mdl-38308646

ABSTRACT

C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.


Subject(s)
C-Peptide , Humans , C-Peptide/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Signal Transduction
2.
Am J Physiol Renal Physiol ; 326(3): F301-F312, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38153850

ABSTRACT

Diabetic kidney disease (DKD) is a serious complication of diabetes affecting millions of people worldwide. Macrophages, a critical immune cell type, are central players in the development and progression of DKD. In this comprehensive review, we delve into the intricate role of macrophages in DKD, examining how they can become polarized into proinflammatory M1 or anti-inflammatory M2 phenotypes. We explore the signaling pathways involved in macrophage recruitment and polarization in the kidneys, including the key cytokines and transcription factors that promote M1 and M2 polarization. In addition, we discuss the latest clinical studies investigating macrophages in DKD and explore the potential of hypoglycemic drugs for modulating macrophage polarization. By gaining a deeper understanding of the mechanisms that regulate macrophage polarization in DKD, we may identify novel therapeutic targets for this debilitating complication of diabetes. This review provides valuable insights into the complex interplay between macrophages and DKD, shedding light on the latest developments in this important area of research. This review aims to enhance understanding of the role that macrophages play in the pathogenesis of DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Humans , Diabetic Nephropathies/metabolism , Signal Transduction , Macrophage Activation , Macrophages/metabolism , Disease Progression , Diabetes Mellitus/metabolism
3.
Mol Med ; 30(1): 80, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858657

ABSTRACT

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a chronic metabolic disease that commonly results from a high-calorie diet and sedentary lifestyle, leading to insulin resistance and glucose homeostasis perturbation. Physical activity is recommended as one first-line treatment in T2DM, but it leads to contrasted results. We hypothesized that, instead of applying standard exercise protocols, the prescription of personalized exercise programs specifically designed to reverse the potential metabolic alterations in skeletal muscle could result in better results. METHODS: To test this hypothesis, we drew the metabolic signature of the fast-twitch quadriceps muscle, based on a combined unbiased NMR spectroscopy and RT-qPCR study, in several T2DM mouse models of different genetic background (129S1/SvImJ, C57Bl/6J), sex and aetiology (high-fat diet (HFD) or HFD/Streptozotocin (STZ) induction or transgenic MKR (FVB-Tg Ckm-IGF1R*K1003R)1Dlr/J) mice. Three selected mouse models with unique muscular metabolic signatures were submitted to three different swimming-based programs, designed to address each metabolic specificity. RESULTS: We found that depending on the genetic background, the sex, and the mode of T2DM induction, specific muscular adaptations occurred, including depressed glycolysis associated with elevated PDK4 expression, shift to ß-oxidation, or deregulation of amino-acid homeostasis. Interestingly, dedicated swimming-based exercises designed to restore specific metabolic alterations in muscle were found optimal in improving systemic T2DM hallmarks, including a significant reduction in insulin resistance, the improvement of glucose homeostasis, and a delay in sensorimotor function alterations. CONCLUSION: The muscle metabolism constitutes an important clue for the design of precision exercises with potential clinical implications for T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2 , Disease Models, Animal , Muscle, Skeletal , Physical Conditioning, Animal , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/genetics , Muscle, Skeletal/metabolism , Mice , Male , Female , Diet, High-Fat/adverse effects , Mice, Inbred C57BL , Insulin Resistance , Metabolome , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Mice, Transgenic , Metabolomics/methods
4.
FASEB J ; 36(10): e22545, 2022 10.
Article in English | MEDLINE | ID: mdl-36094323

ABSTRACT

The kidneys are radiosensitive and dose-limiting organs for radiotherapy (RT) targeting abdominal and paraspinal tumors. Excessive radiation doses to the kidneys ultimately lead to radiation nephropathy. Our prior work unmasked a novel role for the lipid-modifying enzyme, sphingomyelin phosphodiesterase acid-like 3b (SMPDL3b), in regulating the response of renal podocytes to radiation injury. In this study, we investigated the role of SMPDL3b in DNA double-strand breaks (DSBs) repair in vitro and in vivo. We assessed the kinetics of DSBs recognition and repair along with the ATM pathway and nuclear sphingolipid metabolism in wild-type (WT) and SMPDL3b overexpressing (OE) human podocytes. We also assessed the extent of DNA damage repair in SMPDL3b knock-down (KD) human podocytes, and C57BL6 WT and podocyte-specific SMPDL3b-knock out (KO) mice after radiation injury. We found that SMPDL3b overexpression enhanced DSBs recognition and repair through modulating ATM nuclear shuttling. OE podocytes were protected against radiation-induced apoptosis by increasing the phosphorylation of p53 at serine 15 and attenuating subsequent caspase-3 cleavage. SMPDL3b overexpression prevented radiation-induced alterations in nuclear ceramide-1-phosphate (C1P) and ceramide levels. Interestingly, exogenous C1P pretreatment radiosensitized OE podocytes by delaying ATM nuclear foci formation and DSBs repair. On the other hand, SMPDL3b knock-down, in vitro and in vivo, induced a significant delay in DSBs repair. Additionally, increased activation of apoptosis was induced in podocytes of SMPDL3b-KO mice compared to WT mice at 24 h post-irradiation. Together, our results unravel a novel role for SMPDL3b in radiation-induced DNA damage response. The current work suggests that SMPDL3b modulates nuclear sphingolipid metabolism, ATM nuclear shuttling, and DSBs repair.


Subject(s)
Podocytes , Radiation Injuries , Animals , Ceramides/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3 , DNA Breaks, Double-Stranded , Humans , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/metabolism , Radiation Injuries/genetics , Radiation Injuries/metabolism , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism
5.
Cell Mol Life Sci ; 79(9): 502, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36040503

ABSTRACT

Diabetes changes the host microbiota, a condition known as dysbiosis. Dysbiosis is an important factor for the pathogenesis of diabetes and colorectal cancer (CRC). We aimed at identifying the microbial signature associated with diabetes and CRC; and identifying the signaling mechanism altered by dysbiosis and leading to CRC progression in diabetes. MKR mice that can spontaneously develop type 2 diabetes were used. For CRC induction, another subset of mice was treated with azoxymethane and dextran sulfate sodium. To identify the role of microbiota, microbiota-depleted mice were inoculated with fecal microbial transplant from diabetic and CRC mice. Further, a mouse group was treated with probiotics. At the end of the treatment, 16S rRNA sequencing was performed to identify microbiota in the fecal samples. Blood was collected, and colons were harvested for molecular, anatomical, and histological analysis. Our results show that diabetes is associated with a microbial signature characterized by reduction of butyrate-forming bacteria. This dysbiosis is associated with gastrointestinal complications reflected by a reduction in colon lengths. These changes are reversed upon treatment with probiotics, which rectified the observed dysbiosis. Inoculation of control mice with diabetic or cancer microbiota resulted in the development of increased number of polyps. Our data also show that inflammatory cytokines (mainly interleukin (IL)-1ß) and NADPH oxidase (NOX)4 are over-expressed in the colon tissues of diabetic mice. Collectively our data suggest that diabetes is associated with dysbiosis characterized by lower abundance of butyrate-forming bacteria leading to over-expression of IL-1ß and NOX4 leading to gastrointestinal complications and CRC.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Animals , Bacteria/genetics , Butyrates/pharmacology , Carcinogenesis , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Dysbiosis/microbiology , Mice , Mice, Inbred C57BL , NADPH Oxidase 4/genetics , RNA, Ribosomal, 16S
6.
FASEB J ; 34(6): 7915-7926, 2020 06.
Article in English | MEDLINE | ID: mdl-32293077

ABSTRACT

The intracellular molecular pathways involved in radiation-induced nephropathy are still poorly understood. Glomerular endothelial cells are key components of the structure and function of the glomerular filtration barrier but little is known about the mechanisms implicated in their injury and repair. The current study establishes the response of immortalized human glomerular endothelial cells (GEnC) to ionizing radiation (IR). We investigated the role of sphingolipids and the lipid-modifying enzyme sphingomyelin phosphodiesterase acid-like 3b (SMPDL3b) in radiation-induced GEnC damage. After delivering a single dose of radiation, long and very-long-chain ceramide species, and the expression levels of SMPDL3b were elevated. In contrast, levels of ceramide-1-phosphate (C1P) dropped in a time-dependent manner although mRNA and protein levels of ceramide kinase (CERK) remained stable. Treatment with C1P or knocking down SMPDL3b partially restored cell survival and conferred radioprotection. We also report a novel role for the NADPH oxidase enzymes (NOXs), namely NOX1, and NOX-derived reactive oxygen species (ROS) in radiation-induced GEnC damage. Subjecting cultured endothelial cells to radiation was associated with increased NOX activity and superoxide anion generation. Silencing NOX1 using NOX1-specific siRNA mitigated radiation-induced oxidative stress and cellular injury. In addition, we report a novel connection between NOX and SMPDL3b. Treatment with the NOX inhibitor, GKT, decreased radiation-induced cellular injury and restored SMPDL3b basal levels of expression. Our findings indicate the importance of SMPDL3b as a potential therapeutic target in radiation-induced kidney damage.


Subject(s)
Endothelial Cells/metabolism , Kidney Diseases/metabolism , Kidney Glomerulus/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Cell Line , Humans , Kidney Glomerulus/radiation effects , Male , Mice, Inbred C57BL , NADPH Oxidase 1/metabolism , RNA, Messenger/metabolism , Radiation , Reactive Oxygen Species/metabolism , Superoxides/metabolism
7.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672515

ABSTRACT

Immunotherapy is now a recognized treatment option for several types of cancer. However, some cancer patients treated with immune checkpoint inhibitors (ICIs) are subject to immune-related adverse events, including induced diabetes mellitus. The exact role and molecular/genetic action of ICIs in diabetes are still not well understood. Elucidating the underlying mechanisms in a proper fashion would allow better refining of biomarkers that would help diagnose patients at risk of altered immune system homeostasis, but would also hold the potential of new therapeutic options for diabetes. In the present narrative review, we propose to discuss the case of autoimmune diabetes following treatment with ICIs and the role of ICIs in the pathophysiology of diabetes. We also present some scarce available data on interesting potential immune therapies for diabetes.


Subject(s)
Diabetes Mellitus/chemically induced , Diabetes Mellitus/immunology , Immune Checkpoint Inhibitors/adverse effects , T-Lymphocytes/immunology , Animals , B7-H1 Antigen/metabolism , Diabetes Mellitus/pathology , Humans , Immunotherapy , Programmed Cell Death 1 Receptor/metabolism
8.
Int J Mol Sci ; 22(3)2021 Jan 23.
Article in English | MEDLINE | ID: mdl-33498614

ABSTRACT

Oxidative damage by reactive oxygen species (ROS) is one of the main contributors to cell injury and tissue damage in thalassemia patients. Recent studies suggest that ROS generation in non-transfusion-dependent (NTDT) patients occurs as a result of iron overload. Among the different sources of ROS, the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes and cytochrome P450 (CYP450) have been proposed to be major contributors for oxidative stress in several diseases. However, the sources of ROS in patients with NTDT remain poorly understood. In this study, Hbbth3/+ mice, a mouse model for ß-thalassemia, were used. These mice exhibit an unchanged or decreased expression of the major NOX isoforms, NOX1, NOX2 and NOX4, when compared to their C57BL/6 control littermates. However, a significant increase in the protein synthesis of CYP4A and CYP4F was observed in the Hbbth3/+ mice when compared to the C57BL/6 control mice. These changes were paralleled by an increased production of 20-hydroxyeicosatetraenoic acid (20-HETE), a CYP4A and CYP4F metabolite. Furthermore, these changes corroborate with onset of ROS production concomitant with liver injury. To our knowledge, this is the first report indicating that CYP450 4A and 4F-induced 20-HETE production mediates reactive oxygen species overgeneration in Hbbth3/+ mice through an NADPH-dependent pathway.


Subject(s)
Cytochrome P450 Family 4/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Reactive Oxygen Species/metabolism , beta-Thalassemia/metabolism , Animals , Disease Models, Animal , Hepatitis/etiology , Hepatitis/pathology , Iron/metabolism , Isoenzymes/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , beta-Thalassemia/complications , beta-Thalassemia/pathology
9.
Rev Endocr Metab Disord ; 21(4): 451-463, 2020 12.
Article in English | MEDLINE | ID: mdl-32743793

ABSTRACT

In light of the most challenging public health crisis of modern history, COVID-19 mortality continues to rise at an alarming rate. Patients with co-morbidities such as hypertension, cardiovascular disease, and diabetes mellitus (DM) seem to be more prone to severe symptoms and appear to have a higher mortality rate. In this review, we elucidate suggested mechanisms underlying the increased susceptibility of patients with diabetes to infection with SARS-CoV-2 with a more severe COVID-19 disease. The worsened prognosis of COVID-19 patients with DM can be attributed to a facilitated viral uptake assisted by the host's receptor angiotensin-converting enzyme 2 (ACE2). It can also be associated with a higher basal level of pro-inflammatory cytokines present in patients with diabetes, which enables a hyperinflammatory "cytokine storm" in response to the virus. This review also suggests a link between elevated levels of IL-6 and AMPK/mTOR signaling pathway and their role in exacerbating diabetes-induced complications and insulin resistance. If further studied, these findings could help identify novel therapeutic intervention strategies for patients with diabetes comorbid with COVID-19.


Subject(s)
Comorbidity , Coronavirus Infections/immunology , Diabetes Mellitus/immunology , Disease Susceptibility/immunology , Pandemics , Pneumonia, Viral/immunology , COVID-19 , Coronavirus Infections/epidemiology , Diabetes Mellitus/epidemiology , Disease Susceptibility/epidemiology , Humans , Pneumonia, Viral/epidemiology
10.
Clin Sci (Lond) ; 134(4): 403-417, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32095833

ABSTRACT

Diabetic kidney disease is one of the most serious complications of diabetes worldwide and is the leading cause of end-stage renal disease. While research has primarily focused on hyperglycemia as a key player in the pathophysiology of diabetic complications, recently, increasing evidence have underlined the role of adipose inflammation in modulating the development and/or progression of diabetic kidney disease. This review focuses on how adipose inflammation contribute to diabetic kidney disease. Furthermore, it discusses in detail the underlying mechanisms of adipose inflammation, including pro-inflammatory cytokines, oxidative stress, and AMPK/mTOR signaling pathway and critically describes their role in diabetic kidney disease. This in-depth understanding of adipose inflammation and its impact on diabetic kidney disease highlights the need for novel interventions in the treatment of diabetic complications.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipose Tissue/pathology , Inflammation/pathology , Kidney/injuries , NADPH Oxidase 4/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Humans
11.
FASEB J ; 33(12): 14051-14066, 2019 12.
Article in English | MEDLINE | ID: mdl-31661292

ABSTRACT

Cancer was recently annexed to diabetic complications. Furthermore, recent studies suggest that cancer can increase the risk of diabetes. Consequently, diabetes and cancer share many risk factors, but the cellular and molecular pathways correlating diabetes and colon and rectal cancer (CRC) remain far from understood. In this study, we assess the effect of hyperglycemia on cancer cell aggressiveness in human colon epithelial adenocarcinoma cells in vitro and in an experimental animal model of CRC. Our results show that Nox (NADPH oxidase enzyme) 4-induced reactive oxygen species (ROS) production is deregulated in both diabetes and CRC. This is paralleled by inactivation of the AMPK and activation of the mammalian target of rapamycin (mTOR) C1 signaling pathways, resulting in 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) accumulation, induction of DNA damage, and exacerbation of cancer cell aggressiveness, thus contributing to the genomic instability and predisposition to increased tumorigenesis in the diabetic milieu. Pharmacologic activation of AMPK, inhibition of mTORC1, or blockade of Nox4 reduce ROS production, restore the homeostatic signaling of 8-oxoguanine DNA glycosylase/8-oxodG, and lessen the progression of CRC malignancy in a diabetic milieu. Taken together, our results identify the AMPK/mTORC1/Nox4 signaling axis as a molecular switch correlating diabetes and CRC. Modulating this pathway may be a strategic target of therapeutic potential aimed at reversing or slowing the progression of CRC in patients with or without diabetes.-Mroueh, F. M., Noureldein, M., Zeidan, Y. H., Boutary, S., Irani, S. A. M., Eid, S., Haddad, M., Barakat, R., Harb, F., Costantine, J., Kanj, R., Sauleau, E.-A., Ouhtit, A., Azar, S. T., Eid, A. H., Eid, A. A. Unmasking the interplay between mTOR and Nox4: novel insights into the mechanism connecting diabetes and cancer.


Subject(s)
NADPH Oxidase 4/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/pharmacology , Blood Glucose , Caco-2 Cells , DNA Damage , Diabetes Mellitus, Experimental , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADPH Oxidase 4/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Up-Regulation
12.
Int J Mol Sci ; 21(14)2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32674299

ABSTRACT

Diabetic cardiomyopathy (DCM) is a constellation of symptoms consisting of ventricular dysfunction and cardiomyocyte disarray in the presence of diabetes. The exact cause of this type of cardiomyopathy is still unknown; however, several processes involving the mitochondria, such as lipid and glucose metabolism, reactive oxygen species (ROS) production, apoptosis, autophagy and mitochondrial biogenesis have been implicated. In addition, polyphenols have been shown to improve the progression of diabetes. In this review, we discuss some of the mechanisms by which polyphenols, particularly resveratrol, play a role in slowing the progression of DCM. The most important intermediates by which polyphenols exert their protective effect include Bcl-2, UCP2, SIRT-1, AMPK and JNK1. Bcl-2 acts to attenuate apoptosis, UCP2 decreases oxidative stress, SIRT-1 increases mitochondrial biogenesis and decreases oxidative stress, AMPK increases autophagy, and JNK1 decreases apoptosis and increases autophagy. Our dissection of these molecular players aims to provide potential therapeutic targets for the treatment of DCM.


Subject(s)
Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/metabolism , Mitochondria/drug effects , Polyphenols/pharmacology , Polyphenols/therapeutic use , Animals , Apoptosis/drug effects , Humans , Mitochondria/metabolism , Oxidative Stress/drug effects
13.
Bioorg Med Chem Lett ; 29(13): 1580-1585, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31078409

ABSTRACT

Diabetic nephropathy (DN) is one of the most serious complications of diabetes worldwide. It is depicted as the leading cause of end-stage renal disease. Oxidative stress plays a key role in hyperglycemia-induced DN. The preparation and characterization of novel mono-, di-, and trisubstituted-s-triazines endowed with uracil and/or thymine are described in this paper. The synthesis of the title compounds was realized through selective nucleophilic substitution reactions of cyanuric chloride with the corresponding hydrazide nucleobases. In this study, we assessed the effects of these derivatives on the progression of diabetic nephropathy. Our results show that trisubstituted-s-triazines endowed with acylhydrazides attenuate high-glucose induced glomerular mesangial cells proliferation and matrix protein accumulation in vitro. Notably, these derivatives also display anti-oxidative properties. This suggests that the novel trisubstituted-s-triazine derivatives provide renal protection through a reactive oxygen species (ROS)-dependent mechanism. Our data provide evidence that these derivatives may serve as potential therapeutic candidates in the treatment of DN.


Subject(s)
Diabetic Nephropathies/drug therapy , Pyrimidines/therapeutic use , Cell Proliferation , Humans , Mesangial Cells , Pyrimidines/pharmacology , Reactive Oxygen Species
14.
Am J Physiol Renal Physiol ; 315(3): F572-F582, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29767558

ABSTRACT

Nephrotoxicity is a serious side effect for the immunosuppressant drug cyclosporine A(CSA). In this study, we tested the hypothesis that administration of calcium channel blockers such as verapamil or nifedipine ameliorates renal CSA-induced renal dysfunction. Furthermore, our study investigates the roles of inflammatory, oxidative, and fibrotic pathways in CSA-induced renal dysfunction. Six groups of male rats ( n = 6/group) were used and received one of the following treatments for seven consecutive days: vehicle (Cremophor EL ip), CSA (25 mg·kg-1·day-1 ip), verapamil (2 mg·kg-1·day-1 ip), nifedipine (3 mg·kg-1·day-1 ip), CSA in the presence or absence of either verapamil, or nifedipine. Biochemical and histomorphometric analyses showed that rats treated with CSA exhibited clear signs of nephrotoxicity that included 1) proteinuria and elevations in serum creatinine and blood urea nitrogen, 2) mesangial expansion, 3) increases in glomerular and tubular type IV collagen expression, and 4) increases in the glomerulosclerosis and tubulointerstitial fibrosis indices. Although the single administration of nifedipine or verapamil had no significant effect on renal pathology, or its biochemical and physiological function, the concurrent use of either calcium channel blockers significantly and equipotently ameliorated the biochemical, morphological, and functional derangements caused by CSA. More importantly, we report that the oxidative (reactive oxygen species production, NADPH-oxidase activity, and dual oxidase 1/2 levels), fibrotic (transforming growth factor-ß1 expression), and inflammatory (NF-κB expression) manifestations of renal toxicity induced by CSA were significantly reversed upon administration of nifedipine or verapamil. Together, these results highlight the efficacy of calcium channel-blocking agents in attenuating CSA-induced nephrotoxicity and predisposing biochemical and molecular machineries.


Subject(s)
Calcium Channel Blockers/pharmacology , Cyclosporine , Kidney Diseases/prevention & control , Kidney/drug effects , Nifedipine/pharmacology , Verapamil/pharmacology , Animals , Biomarkers/blood , Blood Urea Nitrogen , Collagen Type IV/metabolism , Creatinine/blood , Dual Oxidases/metabolism , Fibrosis , Inflammation Mediators/metabolism , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Male , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Proteinuria/metabolism , Proteinuria/physiopathology , Proteinuria/prevention & control , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
15.
Microb Pathog ; 118: 98-104, 2018 May.
Article in English | MEDLINE | ID: mdl-29548696

ABSTRACT

The gut microbiota plays a substantial role in regulating the host metabolic and immune functions. Dysbiosis, resulting from disruption of gut microbiota, predisposes many morbid pathologies like obesity and its associated comorbidities, diabetes and inflammatory conditions including some types of cancer. There are numerous proposed signaling pathways through which alterations in gut microbiota and its metabolites can disturb the host's normal physiological functions. Interestingly, many of these processes happen to be controlled by the mammalian target of rapamycin (mTOR). The mTOR pathway responds to environmental changes and regulates accordingly many intracellular processes such as transcription, translation, cell growth, cytoskeletal organization and autophagy. In this review, we aim to highlight the cross-talk between the gut microbiota and the mTOR pathway and discuss how this emerging field of research gives a beautiful insight into how the mentioned cross-talk impacts the body's homeostasis thus leading to undesirable complications including obesity, diabetes, colon and pancreatic cancer, immune system malfunctioning and ageing. Although there are a limited number of studies investigating the crosstalk between the gut microbiota and the mTOR pathway, the results obtained so far are enough to elucidate the key role of the mTOR signaling in microbiota-associated metabolic and immune regulations.


Subject(s)
Gastrointestinal Microbiome/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Butyrates , Cellular Senescence , Colonic Neoplasms/complications , Colonic Neoplasms/microbiology , Colonic Neoplasms/physiopathology , Diabetes Complications/microbiology , Diabetes Complications/physiopathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiopathology , Homeostasis , Humans , Immune System/abnormalities , Immunity , Inflammation/microbiology , Metabolic Diseases , Obesity/complications , Obesity/microbiology , Obesity/physiopathology , Pancreatic Neoplasms/complications , Pancreatic Neoplasms/microbiology , Pancreatic Neoplasms/physiopathology , Signal Transduction/immunology , TOR Serine-Threonine Kinases/immunology
16.
Inflamm Res ; 67(2): 191-201, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29085960

ABSTRACT

OBJECTIVE AND DESIGN: The aim of this study is to elucidate TGF-ß1 signaling pathways involved in COX-2 protein induction and modulation of TAU protein phosphorylation in cultured podocytes. MATERIALS, TREATMENT AND METHODS: In vitro cultured immortalized podocytes were stimulated with TGF-ß1 in presence and absence of pharmacologic inhibitors for various signaling pathways and phosphatases. Then, COX-2 protein expression, as well as P38MAPK, AKT and TAU phosphorylation levels were evaluated by western blot analysis. RESULTS: TGF-ß1 induction of COX-2 protein levels was completely blocked by pharmacologic inhibitors of phosphatases, P38 MAPK, or NF-қB pathways. Time course experiments showed that TGF-ß1 activated p38 MAPK after 5 min of stimulation. Interestingly, podocyte co-incubated with TGF-ß1, high glucose and/or PGE2 showed strong increase in p38 MAPK and AKT phosphorylation as well as COX- 2 protein expression levels. Levels of phosphorylated AKT were further reduced and levels of phosphorylated p38 were increased when PGE2 was added to the culture media. Interestingly, selective phosphatases inhibitors completely abrogated PGE2-induced P38 MAPK and TAU phosphorylation. Also, inhibition of phosphatases reversed TGF-ß1-induced COX-2 protein expression either alone or when incubated with high glucose or PGE2. CONCLUSION: These data suggest TGF-ß1 mediates its effect in podocyte through novel signaling mechanisms including phosphatases and TAU protein phosphorylation.


Subject(s)
Cyclooxygenase 2/biosynthesis , Phosphoric Monoester Hydrolases/pharmacology , Podocytes/metabolism , Transforming Growth Factor beta1/pharmacology , tau Proteins/metabolism , Animals , Cells, Cultured , Glucose/pharmacology , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Podocytes/drug effects , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Toxicol Appl Pharmacol ; 334: 110-119, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28899749

ABSTRACT

The endothelium-derived hyperpolarizing factor (EDHF) serves as a back-up mechanism that compensates for reduced nitric oxide (NO)/prostanoids bioavailability. Here we investigated whether (i) under conditions of vascular endothelium dysfunction, the immunosuppressant drug cyclosporine (CSA) upregulates EDHF-dependent renal vasodilations through altering CYP4A/CYP2C signaling, and (ii) calcium channel blockers modulate the CSA/EDHF/CYP interaction. Rats were treated with CSA, verapamil, nifedipine, or their combinations for 7days. Blood pressure (BP) was measured by tail-cuff plethysmography. Kidneys were then isolated, perfused with physiological solution containing L-NAME (NOS inhibitor) and diclofenac (cyclooxygenase inhibitor, DIC), and preconstricted with phenylephrine. CSA (25mgkg-1day-1 for 7days) increased BP and augmented carbachol renal vasodilations. The co-treatment with verapamil (2mgkg-1day-1) or nifedipine (3mgkg-1day-1) abolished CSA hypertension and conversely affected carbachol vasodilations (increases vs. decreases). Infusion of MSPPOH (epoxyeicosatrienoic acids, EETs, inhibitor) reduced carbachol vasodilations in kidneys of all rat groups, suggesting the importance of EETs in these responses. By contrast, 20-Hydroxyeicosatetraenoic Acid (20-HETE) inhibition by HET0016 increased carbachol vasodilations in control rats, an effect that disappeared by CSA treatment, and reappeared in rats treated with CSA/verapamil or CSA/nifedipine. Renal protein expression of CYP2C and CYP4A as well as their vasoactive products (EETs/20-HETE) were increased in CSA-treated rats. Whereas the CYP2C/EETs effects of CSA were abolished by verapamil and intensified by nifedipine, the CYP4A/20-HETE effects were reduced by either CCB. Overall, nifedipine and verapamil blunts CSA hypertension but variably affected concomitantly enhanced EDHF-dependent renal vasodilations and alterations in CYP2C/CYP4A signaling.


Subject(s)
Biological Factors/metabolism , Calcium Channel Blockers/pharmacology , Cyclosporine/pharmacology , Cytochrome P-450 CYP4A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Immunosuppressive Agents/pharmacology , Animals , Calcium Channel Blockers/administration & dosage , Cyclosporine/administration & dosage , Cytochrome P-450 CYP4A/genetics , Cytochrome P-450 Enzyme System/genetics , Drug Interactions , Gene Expression Regulation, Enzymologic/drug effects , Immunosuppressive Agents/administration & dosage , Kidney/blood supply , Male , Nifedipine/administration & dosage , Nifedipine/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , Verapamil/administration & dosage , Verapamil/pharmacology
18.
Int J Mol Sci ; 18(12)2017 Nov 25.
Article in English | MEDLINE | ID: mdl-29186855

ABSTRACT

Sphingolipids, long thought to be passive components of biological membranes with merely a structural role, have proved throughout the past decade to be major players in the pathogenesis of many human diseases. The study and characterization of several genetic disorders like Fabry's and Tay Sachs, where sphingolipid metabolism is disrupted, leading to a systemic array of clinical symptoms, have indeed helped elucidate and appreciate the importance of sphingolipids and their metabolites as active signaling molecules. In addition to being involved in dynamic cellular processes like apoptosis, senescence and differentiation, sphingolipids are implicated in critical physiological functions such as immune responses and pathophysiological conditions like inflammation and insulin resistance. Interestingly, the kidneys are among the most sensitive organ systems to sphingolipid alterations, rendering these molecules and the enzymes involved in their metabolism, promising therapeutic targets for numerous nephropathic complications that stand behind podocyte injury and renal failure.


Subject(s)
Fabry Disease/metabolism , Kidney Diseases/metabolism , Podocytes/metabolism , Sphingolipids/metabolism , Tay-Sachs Disease/metabolism , Animals , Fabry Disease/genetics , Fabry Disease/therapy , Humans , Kidney Diseases/genetics , Kidney Diseases/therapy , Tay-Sachs Disease/genetics , Tay-Sachs Disease/therapy , Translational Research, Biomedical
19.
Heart Fail Rev ; 21(1): 25-47, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26498937

ABSTRACT

Despite dramatic improvements in short-term mortality rates following myocardial infarction (MI), long-term survival for MI patients who progress to heart failure remains poor. MI occurs when the left ventricle (LV) is deprived of oxygen for a sufficient period of time to induce irreversible necrosis of the myocardium. The LV response to MI involves significant tissue, cellular, and molecular level modifications, as well as substantial hemodynamic changes that feedback negatively to amplify the response. Inflammation to remove necrotic myocytes and fibroblast activation to form a scar are key wound healing responses that are highly variable across individuals. Few biomarkers of early remodeling stages are currently clinically adopted. The discovery of underlying pathophysiological mechanisms and associated novel biomarkers has the potential of improving prognostic capability and therapeutic monitoring. Combining these biomarkers with other prominent ones could constitute a powerful diagnostic and prognostic tool that directly reflects the pathophysiological remodeling of the LV. Understanding temporal remodeling at the tissue, cellular, and molecular level and its link to a well-defined set of biomarkers at early stages post-MI is a prerequisite for improving personalized care and devising more successful therapeutic interventions. Here we summarize the integral mechanisms that occur during early cardiac remodeling in the post-MI setting and highlight the most prominent biomarkers for assessing disease progression.

20.
Bioorg Med Chem Lett ; 26(3): 1020-1024, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26733477

ABSTRACT

We report herein the synthesis of a novel series of carbocyclic acylhydrazone derivatives of uracil, thymine and cytosine from the corresponding nucleic bases and their biological activity to treat diabetic nephropathy. Intriguingly, five derivatives significantly reduced high-glucose induced glomerular mesangial cells proliferation and matrix protein accumulation in vitro. The anti-oxidative effects displayed by these molecules suggest that their activity might involve a ROS-dependent mechanism.


Subject(s)
Nucleosides/chemistry , Pyrimidines/chemistry , Actins/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Fibronectins/metabolism , Glucose/pharmacology , Mesangial Cells/cytology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Nucleosides/chemical synthesis , Nucleosides/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Rats , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL