Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Curr Res Physiol ; 7: 100126, 2024.
Article in English | MEDLINE | ID: mdl-38779598

ABSTRACT

Chronic kidney disease (CKD) is a progressive and long-term condition marked by a gradual decline in kidney function. CKD is prevalent among those with conditions such as diabetes mellitus, hypertension, and glomerulonephritis. Affecting over 10% of the global population, CKD stands as a significant cause of morbidity and mortality. Despite substantial advances in understanding CKD pathophysiology and management, there is still a need to explore novel mechanisms and potential therapeutic targets. Urotensin II (UII), a potent vasoactive peptide, has garnered attention for its possible role in the development and progression of CKD. The UII system consists of endogenous ligands UII and UII-related peptide (URP) and their receptor, UT. URP pathophysiology is understudied, but alterations in tissue expression levels of UII and UT and blood or urinary UII concentrations have been linked to cardiovascular and kidney dysfunctions, including systemic hypertension, chronic heart failure, glomerulonephritis, and diabetes. UII gene polymorphisms are associated with increased risk of diabetes. Pharmacological inhibition or genetic ablation of UT mitigated kidney and cardiovascular disease in rodents, making the UII system a potential target for slowing CKD progression. However, a deeper understanding of the UII system's cellular mechanisms in renal and extrarenal organs is essential for comprehending its role in CKD pathophysiology. This review explores the evolving connections between the UII system and CKD, addressing potential mechanisms, therapeutic implications, controversies, and unexplored concepts.

2.
bioRxiv ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38585802

ABSTRACT

Metabolism research is increasingly recognizing the contributions of organelle crosstalk to metabolic regulation. Mitochondria-associated membranes (MAMs), which are structures connecting the mitochondria and endoplasmic reticulum (ER), are critical in a myriad of cellular functions linked to cellular metabolism. MAMs control calcium signaling, mitochondrial transport, redox balance, protein folding/degradation, and in some studies, metabolic health. The possibility that MAMs drive changes in cellular function in individuals with Type 2 Diabetes (T2D) is controversial. Although disruptions in MAMs that change the distance between the mitochondria and ER, MAM protein composition, or disrupt downstream signaling, can perpetuate inflammation, one key trait of T2D. However, the full scope of this structure's role in immune cell health and thus T2D-associated inflammation remains unknown. We show that human immune cell MAM proteins and their associated functions are not altered by T2D and thus unlikely to contribute to metaflammation.

3.
Trends Endocrinol Metab ; 34(10): 583-585, 2023 10.
Article in English | MEDLINE | ID: mdl-37625920

ABSTRACT

Increasing evidence suggests that the brain plays a key role in glucose homeostasis, making it a potential target for the treatment of type 2 diabetes (T2D). Sun et al. recently reported that intracerebroventricular (ICV) administration of a single dose of fibroblast growth factor 4 (FGF4) can induce sustained T2D remission in mouse models in the absence of any risk of hypoglycemia.


Subject(s)
Diabetes Mellitus, Type 2 , Hyperglycemia , Hypoglycemia , Animals , Mice , Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factor 4 , Hyperglycemia/drug therapy , Obesity/drug therapy
4.
Int J Mol Sci ; 24(10)2023 May 15.
Article in English | MEDLINE | ID: mdl-37240109

ABSTRACT

Retinal pigment epithelial (RPE) cell dysfunction is a key driving force of AMD. RPE cells form a metabolic interface between photoreceptors and choriocapillaris, performing essential functions for retinal homeostasis. Through their multiple functions, RPE cells are constantly exposed to oxidative stress, which leads to the accumulation of damaged proteins, lipids, nucleic acids, and cellular organelles, including mitochondria. As miniature chemical engines of the cell, self-replicating mitochondria are heavily implicated in the aging process through a variety of mechanisms. In the eye, mitochondrial dysfunction is strongly associated with several diseases, including age-related macular degeneration (AMD), which is a leading cause of irreversible vision loss in millions of people globally. Aged mitochondria exhibit decreased rates of oxidative phosphorylation, increased reactive oxygen species (ROS) generation, and increased numbers of mitochondrial DNA mutations. Mitochondrial bioenergetics and autophagy decline during aging because of insufficient free radical scavenger systems, the impairment of DNA repair mechanisms, and reductions in mitochondrial turnover. Recent research has uncovered a much more complex role of mitochondrial function and cytosolic protein translation and proteostasis in AMD pathogenesis. The coupling of autophagy and mitochondrial apoptosis modulates the proteostasis and aging processes. This review aims to summarise and provide a perspective on (i) the current evidence of autophagy, proteostasis, and mitochondrial dysfunction in dry AMD; (ii) current in vitro and in vivo disease models relevant to assessing mitochondrial dysfunction in AMD, and their utility in drug screening; and (iii) ongoing clinical trials targeting mitochondrial dysfunction for AMD therapeutics.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Humans , Aged , Retinal Pigment Epithelium/metabolism , Proteostasis , Autophagy/genetics , Oxidative Stress/genetics , Macular Degeneration/pathology , Mitochondria/metabolism
5.
J Ocul Pharmacol Ther ; 39(2): 159-174, 2023 03.
Article in English | MEDLINE | ID: mdl-36791327

ABSTRACT

Purpose: This study evaluated if tauroursodeoxycholic acid (TUDCA) alleviates pro-inflammatory and endoplasmic reticulum (ER) stress-mediated visual deficits in diabetic tie2-TNF transgenic mice via Takeda G protein-coupled receptor 5 (TGR5) receptor signaling. Methods: Adult tie2-TNF transgenic or age-matched C57BL/6J (wildtype, WT) mice were made diabetic and treated subcutaneously with TUDCA. After 4 weeks, visual function, vascular permeability, immunohistology, and molecular analyses were assessed. Human retinal endothelial cells (HRECs) silenced for TGR5, followed by TNF and high glucose (HG) stress-mediated endothelial permeability, and transendothelial migration of activated leukocytes were assessed with TUDCA in vitro. Results: Compared with WT mice, tie2-TNF mice showed a decreased visual function correlated with a decrease in protein kinase C α (PKCα) in rod bipolar cells, and increased vascular permeability was further exacerbated in diabetic-tie2-TNF mice. Conversely, TUDCA alleviated these changes in diabetic mice. An increase in inflammation and ER stress in retina coincided with an increase in TGR5 expression in diabetic tie2-TNF mice that decreased with TUDCA. In vitro, HRECs exposed to TNF+HG demonstrated >2-fold increase in TGR5 expression, a 3-fold increase in leukocyte transmigration with a concomitant increase in permeability. Although TUDCA reversed these effects, HRECs silenced for TGR5 and challenged with TUDCA or TGR5 agonist failed to reverse the TNF+HG induced effects. Conclusions: Our data suggest that TUDCA will serve as an excellent therapeutic agent for diabetic complications addressing both vascular and neurodegenerative changes in the retina. Perturbation of the TGR5 receptor in the retina might play a role in linking retinal ER stress to neurovascular dysfunction in diabetic retinopathy.


Subject(s)
Diabetes Mellitus, Experimental , Mice , Animals , Humans , Mice, Transgenic , Diabetes Mellitus, Experimental/drug therapy , Endothelial Cells/metabolism , Mice, Inbred C57BL , Endoplasmic Reticulum Stress/physiology
6.
Nat Rev Endocrinol ; 18(1): 23-42, 2022 01.
Article in English | MEDLINE | ID: mdl-34703027

ABSTRACT

Obesity and type 2 diabetes mellitus (T2DM) are increasing in prevalence owing to decreases in physical activity levels and a shift to diets that include addictive and/or high-calorie foods. These changes are associated with the adoption of modern lifestyles and the presence of an obesogenic environment, which have resulted in alterations to metabolism, adaptive immunity and endocrine regulation. The size and quality of adipose tissue depots in obesity, including the adipose tissue immune compartment, are critical determinants of overall health. In obesity, chronic low-grade inflammation can occur in adipose tissue that can progress to systemic inflammation; this inflammation contributes to the development of insulin resistance, T2DM and other comorbidities. An improved understanding of adaptive immune cell dysregulation that occurs during obesity and its associated metabolic comorbidities, with an appreciation of sex differences, will be critical for repurposing or developing immunomodulatory therapies to treat obesity and/or T2DM-associated inflammation. This Review critically discusses how activation and metabolic reprogramming of lymphocytes, that is, T cells and B cells, triggers the onset, development and progression of obesity and T2DM. We also consider the role of immunity in under-appreciated comorbidities of obesity and/or T2DM, such as oral cavity inflammation, neuroinflammation in Alzheimer disease and gut microbiome dysbiosis. Finally, we discuss previous clinical trials of anti-inflammatory medications in T2DM and consider the path forward.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Insulin Resistance , Adaptive Immunity , Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Inflammation , Male , Obesity/metabolism
7.
Stem Cell Res Ther ; 10(1): 318, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690344

ABSTRACT

BACKGROUND: Retinal inflammation affecting the neurovascular unit may play a role in the development of visual deficits following mild traumatic brain injury (mTBI). We have shown that concentrated conditioned media from adipose tissue-derived mesenchymal stem cells (ASC-CCM) can limit retinal damage from blast injury and improve visual function. In this study, we addressed the hypothesis that TNFα-stimulated gene-6 (TSG-6), an anti-inflammatory protein released by mesenchymal cells, mediates the observed therapeutic potential of ASCs via neurovascular modulation. METHODS: About 12-week-old C57Bl/6 mice were subjected to 50-psi air pulse on the left side of the head overlying the forebrain resulting in an mTBI. Age-matched sham blast mice served as control. About 1 µl of ASC-CCM (siControl-ASC-CCM) or TSG-6 knockdown ASC-CCM (siTSG-6-ASC-CCM) was delivered intravitreally into both eyes. One month following injection, the ocular function was assessed followed by molecular and immunohistological analysis. In vitro, mouse microglial cells were used to evaluate the anti-inflammatory effect of ASC-CCM. Efficacy of ASC-CCM in normalizing retinal vascular permeability was assessed using trans-endothelial resistance (TER) and VE-cadherin expression in the presence of TNFα (1 ng/ml). RESULTS: We show that intravitreal injection of ASC-CCM (siControl-ASC-CCM) but not the TSG-6 knockdown ASC-CCM (siTSG-6-ASC-CCM) mitigates the loss of visual acuity and contrast sensitivity, retinal expression of genes associated with microglial and endothelial activation, and retinal GFAP immunoreactivity at 4 weeks after blast injury. In vitro, siControl-ASC-CCM but not the siTSG-6-ASC-CCM not only suppressed microglial activation and STAT3 phosphorylation but also protected against TNFα-induced endothelial permeability as measured by transendothelial electrical resistance and decreased STAT3 phosphorylation. CONCLUSIONS: Our findings suggest that ASCs respond to an inflammatory milieu by secreting higher levels of TSG-6 that mediates the resolution of the inflammatory cascade on multiple cell types and correlates with the therapeutic potency of the ASC-CCM. These results expand our understanding of innate mesenchymal cell function and confirm the importance of considering methods to increase the production of key analytes such as TSG-6 if mesenchymal stem cell secretome-derived biologics are to be developed as a treatment solution against the traumatic effects of blast injuries and other neurovascular inflammatory conditions of the retina.


Subject(s)
Adipose Tissue/cytology , Brain Injuries, Traumatic/physiopathology , Brain Injuries, Traumatic/therapy , Cell Adhesion Molecules/metabolism , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/drug effects , Neovascularization, Physiologic/drug effects , Vision, Ocular/drug effects , Animals , Cell Shape/drug effects , Cytokines/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium/drug effects , Endothelium/pathology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Inflammation/genetics , Inflammation/pathology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Models, Biological , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/drug effects , Retina/pathology , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/toxicity
8.
Sci Rep ; 9(1): 10783, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31346222

ABSTRACT

Increased O-GlcNAcylation, a well-known post-translational modification of proteins causally linked to various detrimental cellular functions in pathological conditions including diabetic retinopathy (DR). Previously we have shown that endothelial activation induced by inflammation and hyperglycemia results in the endoplasmic reticulum (ER) stress-mediated intercellular junction alterations accompanied by visual deficits in a tie2-TNF-α transgenic mouse model. In this study, we tested the hypothesis that increased ER stress via O-GlcNAcylation of VE-Cadherin likely contribute to endothelial permeability. We show that ER stress leads to GRP78 translocation to the plasma membrane, increased O-GlcNAcylation of proteins, particularly VE-Cadherin resulting in a defective complex partnering leading to the loss of retinal endothelial barrier integrity and increased transendothelial migration of monocytes. We further show an association of GRP78 with the VE-Cadherin under these conditions. Interestingly, cells exposed to ER stress inhibitor, tauroursodeoxycholic acid partially mitigated all these effects. Our findings suggest an essential role for ER stress and O-GlcNAcylation in altering the endothelial barrier function and reveal a potential therapeutic target in the treatment of DR.


Subject(s)
Antigens, CD/metabolism , Blood-Retinal Barrier/metabolism , Cadherins/metabolism , Capillary Permeability , Endoplasmic Reticulum Stress , Endothelial Cells/metabolism , Heat-Shock Proteins/metabolism , Blood-Retinal Barrier/cytology , Cell Membrane/metabolism , Cell Movement , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Glycosylation , Humans , Monocytes/physiology , Protein Transport , Taurochenodeoxycholic Acid/pharmacology
9.
Stem Cell Res Ther ; 9(1): 322, 2018 11 21.
Article in English | MEDLINE | ID: mdl-30463601

ABSTRACT

BACKGROUND: Early-stage diabetic retinopathy (DR) is characterized by neurovascular defects. In this study, we hypothesized that human adipose-derived stem cells (ASCs) positive for the pericyte marker CD140b, or their secreted paracrine factors, therapeutically rescue early-stage DR features in an Ins2Akita mouse model. METHODS: Ins2Akita mice at 24 weeks of age received intravitreal injections of CD140b-positive ASCs (1000 cells/1 µL) or 20× conditioned media from cytokine-primed ASCs (ASC-CM, 1 µL). Age-matched wildtype mice that received saline served as controls. Visual function experiments and histological analyses were performed 3 weeks post intravitreal injection. Biochemical and molecular analyses assessed the ASC-CM composition and its biological effects. RESULTS: Three weeks post-injection, Ins2Akita mice that received ASCs had ameliorated decreased b-wave amplitudes and vascular leakage but failed to improve visual acuity, whereas Ins2Akita mice that received ASC-CM demonstrated amelioration of all aforementioned visual deficits. The ASC-CM group demonstrated partial amelioration of retinal GFAP immunoreactivity and DR-related gene expression but the ASC group did not. While Ins2Akita mice that received ASCs exhibited occasional (1 in 8) hemorrhagic retinas, mice that received ASC-CM had no adverse complications. In vitro, ASC-CM protected against TNFα-induced retinal endothelial permeability as measured by transendothelial electrical resistance. Biochemical and molecular analyses demonstrated several anti-inflammatory proteins including TSG-6 being highly expressed in cytokine-primed ASC-CM. CONCLUSIONS: ASCs or their secreted factors mitigate retinal complications of diabetes in the Ins2Akita model. Further investigation is warranted to determine whether ASCs or their secreted factors are safe and effective therapeutic modalities long-term as current locally delivered therapies fail to effectively mitigate the progression of early-stage DR. Nonetheless, our study sheds new light on the therapeutic mechanisms of adult stem cells, with implications for assessing relative risks/benefits of experimental regenerative therapies for vision loss.


Subject(s)
Adipose Tissue/cytology , Diabetes Mellitus, Experimental/complications , Diabetic Retinopathy/therapy , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cell Transplantation , Adipose Tissue/metabolism , Animals , Antigens, Surface/chemistry , Antigens, Surface/therapeutic use , Culture Media, Serum-Free/chemistry , Culture Media, Serum-Free/pharmacology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Humans , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Thrombomodulin
10.
Exp Biol Med (Maywood) ; 243(12): 976-984, 2018 08.
Article in English | MEDLINE | ID: mdl-30114984

ABSTRACT

Stress-associated premature senescence plays a major role in retinal diseases. In this study, we investigated the relationship between endothelial dysfunction, endoplasmic reticulum (ER) stress, and cellular senescence in the development of retinal dysfunction. We tested the hypothesis that constant endothelial activation by transmembrane tumor necrosis factor-α (tmTNF-α) exacerbates age-induced visual deficits via senescence-mediated ER stress in this model. To address this, we employed a mouse model of chronic vascular activation using endothelial-specific TNF-α-expressing (tie2-TNF) mice at 5 and 10 months of age. Visual deficits were exhibited by tie2-TNF mice at both 5 months and 10 months of age, with the older mice showing statistically significant loss of visual acuity compared with tie2-TNF mice at age 5 months. The neural defects, as measured by electroretinogram (ERG), also followed a similar trend in an age-dependent fashion, with 10-month-old tie2-TNF mice showing the greatest decrease in "b" wave amplitude at 25 cd.s.m2 compared with age-matched wildtype (WT) mice and five-month-old tie2-TNF mice. While gene and protein expression from the whole retinal extracts demonstrated increased inflammatory (Icam1, Ccl2), stress-associated premature senescence (p16, p21, p53), and ER stress (Grp78, p-Ire1α, Chop) markers in five-month-old tie2-TNF mice compared with five-month-old WT mice, a further increase was seen in 10-month-old tie2-TNF mice. Our data demonstrate that tie2-TNF mice exhibit age-associated increases in visual deficits, and these data suggest that inflammatory endothelial activation is at least partly at play. Given the correlation of increased premature senescence and ER stress in an age-dependent fashion, with the loss of visual functions and increased endothelial activation, our data suggest a possible self-enhanced loop of unfolded protein response pathways and senescence in propagating neurovascular defects in this model. Impact statement Vision loss in most retinal diseases affects the quality of life of working age adults. Using a novel animal model that displays constant endothelial activation by tmTNF-α, our results demonstrate exacerbated age-induced visual deficits via premature senescence-mediated ER stress. We have compared mice of 5 and 10 months of age, with highly relevant human equivalencies of approximately 35- and 50-year-old patients, representing mature adult and middle-aged subjects, respectively. Our studies suggest a possible role for a self-enhanced loop of ER stress pathways and senescence in the propagation of retinal neurovascular defects, under conditions of constant endothelial activation induced by tmTNF-α signaling.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Receptor, TIE-2/genetics , Tumor Necrosis Factor-alpha/metabolism , Vision Disorders/genetics , Vision, Ocular/genetics , Animals , Cells, Cultured , Cellular Senescence , Electroretinography , Endoplasmic Reticulum Chaperone BiP , Endothelial Cells/cytology , Female , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reflex , Signal Transduction
11.
Int J Mol Sci ; 19(7)2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29997321

ABSTRACT

Blast concussions are a common injury sustained in military combat today. Inflammation due to microglial polarization can drive the development of visual defects following blast injuries. In this study, we assessed whether anti-inflammatory factors released by the mesenchymal stem cells derived from adipose tissue (adipose stem cells, ASC) can limit retinal tissue damage and improve visual function in a mouse model of visual deficits following mild traumatic brain injury. We show that intravitreal injection of 1 µL of ASC concentrated conditioned medium from cells pre-stimulated with inflammatory cytokines (ASC-CCM) mitigates loss of visual acuity and contrast sensitivity four weeks post blast injury. Moreover, blast mice showed increased retinal expression of genes associated with microglial activation and inflammation by molecular analyses, retinal glial fibrillary acidic protein (GFAP) immunoreactivity, and increased loss of ganglion cells. Interestingly, blast mice that received ASC-CCM improved in all parameters above. In vitro, ASC-CCM not only suppressed microglial activation but also protected against Tumor necrosis alpha (TNFα) induced endothelial permeability as measured by transendothelial electrical resistance. Biochemical and molecular analyses demonstrate TSG-6 is highly expressed in ASC-CCM from cells pre-stimulated with TNFα and IFNγ but not from unstimulated cells. Our findings suggest that ASC-CCM mitigates visual deficits of the blast injury through their anti-inflammatory properties on activated pro-inflammatory microglia and endothelial cells. A regenerative therapy for immediate delivery at the time of injury may provide a practical and cost-effective solution against the traumatic effects of blast injuries to the retina.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Blast Injuries/complications , Brain Concussion/etiology , Culture Media, Conditioned/chemistry , Mesenchymal Stem Cells/metabolism , Retinitis/drug therapy , Vision Disorders/drug therapy , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Brain Concussion/complications , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Intravitreal Injections , Male , Mesenchymal Stem Cells/cytology , Mice , Microglia/drug effects , Microglia/metabolism , Retinitis/etiology , Vision Disorders/etiology
12.
J Cell Biochem ; 119(10): 8460-8471, 2018 11.
Article in English | MEDLINE | ID: mdl-30054947

ABSTRACT

Diabetic retinopathy (DR) is the leading cause of vision loss among working-age adults. The interplay between hyperglycemia and endothelial activation in inducing endoplasmic reticulum (ER) stress pathways and visual deficits in DR is not fully understood. To address this, we used a mouse model of chronic vascular activation using endothelial-specific tumor necrosis factor-α (TNF-α)-expressing (tie2-TNF) mice to induce diabetes with streptozotocin. At 4 weeks post streptozotocin, a significant 2-fold to 10-fold increase in retinal neurovascular inflammatory gene transcript response in tie2-TNF mice was further increased in diabetic tie2-TNF mice. A decrease in visual acuity and scotopic b-wave amplitude in tie2-TNF mice was further accentuated in diabetic tie2-TNF mice and these changes correlated with a multi-fold increase in retinal ER stress markers and a reduction in adherens junctions. Cultured retinal endothelial cells showed a significant decrease in trans-endothelial resistance as well as VE-cadherin expression under TNF-α and high glucose stress. These changes were partly rescued by tauroursodeoxycholic acid, a potent ER stress inhibitor. Taken together, constant endothelial activation induced by TNF-α further exacerbated by hyperglycemia results in activation of ER stress and chronic proinflammation in a feed forward loop ultimately resulting in endothelial junction protein alterations leading to visual deficits in the retina. Inhibition of ER stress and endothelial activation may prove to be a novel therapeutic target in DR.


Subject(s)
Diabetic Retinopathy/metabolism , Endoplasmic Reticulum Stress/physiology , Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Analysis of Variance , Animals , Cell Line , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Electroretinography , Gene Expression , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, TIE-2/genetics , Retina/pathology , Streptozocin , Visual Acuity/physiology
13.
Curr Pharm Des ; 24(40): 4742-4754, 2018.
Article in English | MEDLINE | ID: mdl-30644342

ABSTRACT

BACKGROUND: The eye is considered as a window of the disease, and a better understanding of neurodegenerative changes in the eye may help diagnose and manage neurodegenerative diseases including the diseases of brain, heart, kidney and liver. In the eye, the blood retinal barrier (BRB] is maintained by a combination of endothelial cells, pericytes, and glia. This BRB integrity is fundamental to the physiology of retinal cellular function and accurate vision. The role of endothelial dysfunction as a consequence of endothelial activation in the initiation and prolongation of neurovascular diseases of the retina is emerging. METHODS: The observations made in this article are a result of our research over the years in the subject matter and also based on a literature search using PubMed with keywords including but not limited to endothelial, permeability, oxidative stress, ROS, TNF-α, retina, injury, and neurodegeneration. Several studies were identified that fulfilled the inclusion criteria. Overall, published studies support an association between endothelial activation, inflammation and oxidative stress in retinal diseases. Although the selection of specific endothelial activation biomarkers in the retina is less clear, there is an increased association between inflammation in the severity of diabetic retinopathy. Studies in other clinically relevant studies demonstrated a strong association of endothelial activation to alterations in mitochondrial respiratory chain complexes, pericyte integrity, microglial activation, neutrophil extracellular traps and elevated plasma concentrations of TNF-α. CONCLUSION: The compromise in BRB as a consequence of the neurovascular unit in the retinal tissue has gained a lot of attention and studies addressing these should result in a better understanding of the pathophysiology of retinal diseases. Although there are no specific retinal markers of endothelial activation and inflammation, future studies using specific models that display endothelial activation, inflammation and oxidative stress likely yield better understanding on the cause or effect relationship of endothelial activation in retinal diseases.


Subject(s)
Endothelial Cells/metabolism , Nervous System/metabolism , Oxidative Stress , Retina/metabolism , Animals , Blood-Retinal Barrier/metabolism , Humans , Inflammation/metabolism
14.
Clin Epigenetics ; 8: 125, 2016.
Article in English | MEDLINE | ID: mdl-27904654

ABSTRACT

BACKGROUND: A role of proinflammation has been implicated in the pathogenesis of diabetes, but the up-stream regulatory signals and molecular signatures are poorly understood. While histone modifications such as changes in histone deacetylase (HDAC) are emerging as novel epigenetic biomarkers, there is lack of studies to demonstrate their clinical relevance in diabetes. Therefore, we investigated the extent of HDAC machinery and inflammatory signals in peripheral blood mononuclear cells (PBMCs) from patients with type 2 diabetes mellitus (T2DM) compared to control subjects. RESULTS: HDAC3 activity was significantly (p < 0.05) increased in patients with T2DM compared to control subjects. While subtypes of HDACs were differentially expressed at their transcriptional levels in patients with type 2 diabetes, the most prominent observation is the significantly (p < 0.05) elevated messenger RNA (mRNA) levels of HDAC3. Expression levels of Sirt1 which represents the class III HDAC were decreased significantly in T2DM (p < 0.05). Plasma levels of both TNF-α and IL-6 were significantly higher (p < 0.05) in patients with type 2 diabetes compared to control subjects. Among the proinflammatory mediators, the mRNA expression of MCP-1, IL1-ß, NFκB, TLR2, and TLR4 were also significantly (p < 0.05) increased in T2DM. Transcriptional levels of DBC1 (deleted in breast cancer 1, which is a negative regulator of HDAC3) were seen significantly reduced in PBMCs from T2DM. Interestingly, HDAC3 activity/HDAC3 mRNA levels positively correlated to proinflammation, poor glycemic control, and insulin resistance. CONCLUSIONS: Striking message from this study is that while looking for anti-inflammatory strategies and drugs with novel mode of action for T2DM, discovering and designing specific inhibitors targeted to HDAC3 appears promising.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytokines/blood , Diabetes Mellitus, Type 2/metabolism , Histone Deacetylases/metabolism , Insulin Resistance , Sirtuin 1/genetics , Adult , Body Mass Index , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Epigenesis, Genetic , Female , Gene Expression Regulation , Histone Deacetylases/blood , Histone Deacetylases/genetics , Humans , Male , Middle Aged
15.
J Leukoc Biol ; 98(4): 615-22, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26123743

ABSTRACT

The mechanism of perturbed immune function in patients with T2DM is poorly understood. Recent studies imply a role for ER stress in linking immune-system alterations and metabolism. Here, we investigated whether ER stress markers and its downstream effector signals are altered in patients with type 2 diabetes along with proinflammatory augmentation. In our study, gene and protein expression of ER stress markers (GRP-78, PERK, IRE1α, ATF6, XBP-1 and CHOP) was elevated significantly (P < 0.05) in PBMCs from T2DM patients compared with control subjects. The mRNA expression of both the proinflammatory cytokines (TNF-α and IL-6) and oxidative stress markers (p22(phox), TXNIP, and TRPC-6; P < 0.05) was also increased in PBMCs from patients with T2DM. SOCS3 mRNA expression was reduced significantly (P < 0.05) in diabetes patients. mRNA expression of most of the ER stress markers from PBMCs correlated significantly and positively with poor glycemic control, dyslipidemia, IR, and inflammatory and oxidative stress markers. Chronic ER stress in PBMCs from patients with T2DM was evident from the increased caspase-3 activity (P < 0.01), which is an executioner of apoptosis. Along with an impairment of miR-146a levels, the downstream targets of miR-146a, viz., IRAK1 and TRAF6 mRNA levels, were also elevated significantly (P < 0.01) in patients with T2DM. There was an inverse relationship among miR-146a levels and ER stress markers, inflammatory markers, and glycemic control. We demonstrate evidence of increased ER stress markers with impaired miR-146a levels and increased proinflammatory signals in patients with type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress/immunology , Adult , Biomarkers/blood , Cytokines/biosynthesis , Female , Humans , Immunoprecipitation , Male , MicroRNAs/biosynthesis , Middle Aged , Oxidative Stress/physiology , Real-Time Polymerase Chain Reaction
16.
Mol Cell Biochem ; 404(1-2): 271-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25776571

ABSTRACT

Endoplasmic reticulum (ER) stress is emerging as a unifying paradigm and one of the underlying mechanisms in the genesis of diabetes and its complications. While this has prompted the development of ER stress inhibitors, there is a limitation in monitoring of ER stress in vitro and in vivo by reliable methodologies. We validated the secreted alkaline phosphatase (SEAP) activity as a surrogate marker of ER stress in mouse ß-TC6 cells exposed to glucolipotoxicity or tunicamycin and studied insulin secretion along with alterations in ER stress markers. SEAP activity assay was measured using the Great EscAPe SEAP kit, insulin levels were determined by Mercodia reagents and mRNA expression of ER stress markers was quantified by real-time PCR. SEAP activity in ß-cells was significantly decreased (indicating increased ER stress) on exposure either to glucolipotoxicity or tunicamycin. This was accompanied by an increased mRNA expression of ER stress markers (GRP-78, PERK, IRE1α, ATF6, XBP-1, and CHOP) and decreased insulin secretion. Treating the cells with phenylbutyric acid normalized SEAP activity, decreased mRNA expression of ER stress markers and improved insulin secretion. Interestingly, cells exposed to different classes of anti-diabetes agents or compounds such as resveratrol resisted ER stress. Methylglyoxal also induces ER stress and this was counteracted by aminoguanidine. Out study demonstrates SEAP activity as a novel ER stress monitoring assay to investigate the therapeutic value of agents with ER stress inhibitory potential. Future studies should focus on the exercise of adopting this reporter assay for high-throughput screening mode of drug discovery.


Subject(s)
Alkaline Phosphatase/biosynthesis , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum/genetics , Alkaline Phosphatase/genetics , Animals , Biomarkers/metabolism , Cell Line , Endoplasmic Reticulum/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Insulin/metabolism , Insulin Secretion , Mice , Phenylbutyrates/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tunicamycin/toxicity
17.
Exp Diabetes Res ; 2012: 356487, 2012.
Article in English | MEDLINE | ID: mdl-22550476

ABSTRACT

Chronic ER stress is emerging as a trigger that imbalances a number of systemic and arterial-wall factors and promote atherosclerosis. Macrophage apoptosis within advanced atherosclerotic lesions is also known to increase the risk of atherothrombotic disease. We hypothesize that glucolipotoxicity might mediate monocyte activation and apoptosis through ER stress. Therefore, the aims of this study are (a) to investigate whether glucolipotoxicity could impose ER stress and apoptosis in THP-1 human monocytes and (b) to investigate whether 4-Phenyl butyric acid (PBA), a chemical chaperone could resist the glucolipotoxicity-induced ER stress and apoptosis. Cells subjected to either glucolipotoxicity or tunicamycin exhibited increased ROS generation, gene and protein (PERK, GRP-78, IRE1α, and CHOP) expression of ER stress markers. In addition, these cells showed increased TRPC-6 channel expression and apoptosis as revealed by DNA damage and increased caspase-3 activity. While glucolipotoxicity/tunicamycin increased oxidative stress, ER stress, mRNA expression of TRPC-6, and programmed the THP-1 monocytes towards apoptosis, all these molecular perturbations were resisted by PBA. Since ER stress is one of the underlying causes of monocyte dysfunction in diabetes and atherosclerosis, our study emphasize that chemical chaperones such as PBA could alleviate ER stress and have potential to become novel therapeutics.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Monocytes/drug effects , Phenylbutyrates/pharmacology , Caspase 3/metabolism , Cell Line , Cells, Cultured , DNA Damage/drug effects , Humans , Monocytes/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
18.
Indian J Clin Biochem ; 25(2): 111-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-23105896

ABSTRACT

The endoplasmic reticulum (ER) is a cellular compartment responsible for multiple important cellular functions including the biosynthesis and folding of newly synthesized proteins destined for secretion, such as insulin. A myriad of pathological and physiological factors perturb ER function and cause dysregulation of ER homeostasis, leading to ER stress. Accumulating evidence suggests that ER stress plays a role in the pathogenesis of diabetes, contributing to pancreatic ß-cell loss and insulin resistance. ER stress may also link obesity, inflammation and insulin resistance in type 2 diabetes. In this review, we address the transition from physiology to pathology, namely how and why the physiological UPR evolves to a proapoptotic ER stress response in diabetes and its complications. Special attention was given to elucidate how ER stress could explain some of the 'clinical paradoxes' such as secondary sulfonylurea failure, initial worsening of retinopathy during tight glycemic control, insulin resistance induced by protease inhibitors and other clinically relevant observations.

SELECTION OF CITATIONS
SEARCH DETAIL
...