Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 29(1): 103819, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37940034

ABSTRACT

Mitochondria are the primary organelles of cells involved in various physiochemical and biochemical processes. Owing to their crucial role in cellular metabolism, mitochondria are favored therapeutic targets for the treatment and prevention of cancers. Recently, there has been growing interest in the use of mitochondria-specific functional nanoparticles for targeted delivery of therapeutic agents to these organelles. Among several nanosystems, liposomes have garnered considerable attention owing to their exceptional drug delivery capabilities, biocompatibility, biodegradability, ease of manufacturing and established regulatory guidelines for market approval. In this context, the present review provides a brief insight into the association between mitochondria and tumor formation and advantages of mitochondrial targeting in cancer therapy. Furthermore, it discusses mitochondria-targeting functional liposomes for the treatment of various cancers, such as breast, lung, colon, among others.


Subject(s)
Liposomes , Neoplasms , Humans , Liposomes/therapeutic use , Mitochondria/metabolism , Drug Delivery Systems , Neoplasms/drug therapy , Neoplasms/metabolism , Lung/metabolism
2.
Int J Mol Sci ; 24(14)2023 Jul 11.
Article in English | MEDLINE | ID: mdl-37511060

ABSTRACT

Adipocytes play a critical role in maintaining a healthy systemic metabolism by storing and releasing energy in the form of fat and helping to regulate glucose and lipid levels in the body. Adipogenesis is the process through which pre-adipocytes are differentiated into mature adipocytes. It is a complex process involving various transcription factors and signaling pathways. The dysregulation of adipogenesis has been implicated in the development of obesity and metabolic disorders. Therefore, understanding the mechanisms that regulate adipogenesis and the factors that contribute to its dysregulation may provide insights into the prevention and treatment of these conditions. RNA-binding motif single-stranded interacting protein 1 (RBMS1) is a protein that binds to RNA and plays a critical role in various cellular processes such as alternative splicing, mRNA stability, and translation. RBMS1 polymorphism has been shown to be associated with obesity and type 2 diabetes, but the role of RBMS1 in adipose metabolism and adipogenesis is not known. We show that RBMS1 is highly expressed during the early phase of the differentiation of the murine adipocyte cell line 3T3-L1 and is significantly upregulated in the adipose tissue depots and adipocytes of high-fat-fed mice, implying a possible role in adipogenesis and adipose metabolism. Knockdown of RBMS1 in pre-adipocytes impacted the differentiation process and reduced the expression of some of the key adipogenic markers. Transcriptomic and proteomic analysis indicated that RBMS1 depletion affected the expression of several genes involved in major metabolic processes, including carbohydrate and lipid metabolism. Our findings imply that RBMS1 plays an important role in adipocyte metabolism and may offer novel therapeutic opportunity for metabolic disorders such as obesity and type 2 diabetes.


Subject(s)
Adipocytes , Adipogenesis , Animals , Mice , 3T3-L1 Cells , Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Lipid Metabolism/genetics , Obesity/metabolism , Proteomics , Transcriptome
3.
Int J Pharm ; 643: 123276, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37516217

ABSTRACT

Breast cancer is the most prevalent type of cancer worldwide,particularly among women, with substantial side effects after therapy. Despite the availability of numerous therapeutic approaches, particularly chemotherapy, the survival rates for breast cancer have declined over time. The therapies currently utilized for breast cancer treatment do not specifically target cancerous cells, resulting in significant adverse effects and potential harm to healthy cells alongside the cancer cells. As a result, nanoparticle-based drug delivery systems have emerged. Among various types of nanoparticles, natural polysaccharide-based nanoparticles have gained significant attention due to their ability to precisely control the drug release and achieve targeted drug delivery. Moreover, polysaccharides are biocompatible, biodegradable, easily modifiable, and renewable, which makes them a unique material for nanoformulation. In recent years, dextran and its derivatives have gained much interest in the field of breast cancer therapy. Dextran is a hydrophilic polysaccharide composed of a main chain formed by α-1,6 linked glucopyranoside residues and a side chain composed of residues linked in α-1,2/3/4 positions. Different dextran-antitumor medication conjugates enhancethe efficacy of anticancer agents. With this context, the present review provides brief insights into dextran and its modification. Further, it meticulously discusses the role of dextran-based nanoparticles in breast cancer therapy and imaging, followed by snippets on their toxicity. Lastly, it presents clinical trials and future perspectives of dextran-based nanoparticles in breast cancer treatment.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Nanoparticles , Female , Humans , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Dextrans/chemistry , Drug Delivery Systems , Nanoparticles/chemistry , Drug Carriers/chemistry
4.
Membranes (Basel) ; 13(3)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36984730

ABSTRACT

Over the past few years, pharmaceutical and biomedical areas have made the most astounding accomplishments in the field of medicine, diagnostics and drug delivery. Nanotechnology-based tools have played a major role in this. The implementation of this multifaceted nanotechnology concept encourages the advancement of innovative strategies and materials for improving patient compliance. The plausible usage of nanotechnology in drug delivery prompts an extension of lipid-based nanocarriers with a special reference to barriers such as the skin and blood-brain barrier (BBB) that have been discussed in the given manuscript. The limited permeability of these two intriguing biological barriers restricts the penetration of active moieties through the skin and brain, resulting in futile outcomes in several related ailments. Lipid-based nanocarriers provide a possible solution to this problem by facilitating the penetration of drugs across these obstacles, which leads to improvements in their effectiveness. A special emphasis in this review is placed on the composition, mechanism of penetration and recent applications of these carriers. It also includes recent research and the latest findings in the form of patents and clinical trials in this field. The presented data demonstrate the capability of these carriers as potential drug delivery systems across the skin (referred to as topical, dermal and transdermal delivery) as well as to the brain, which can be exploited further for the development of safe and efficacious products.

5.
Biology (Basel) ; 12(3)2023 Mar 09.
Article in English | MEDLINE | ID: mdl-36979108

ABSTRACT

Heat stroke is among the most hazardous hyperthermia-related illnesses and an emerging threat to humans from climate change. Acute brain injury and long-lasting brain damage are the hallmarks of this condition. Hyperthermic neurological manifestations are remarkable for their damage correlation with stress amplitude and long-term persistence. Hyperthermia-induced protein unfolding, and nonspecific aggregation accumulation have neurotoxic effects and contribute to the pathogenesis of brain damage in heat stroke. Therefore, we generated heat-induced, dose-responsive extreme and mild proteotoxic stress models in medulloblastoma [Daoy] and neuroblastoma [SH-SY5Y] and differentiated SH-SY5Y neuronal cells. We show that heat-induced protein aggregation is associated with reduced cell proliferation and viability. Higher protein aggregation in differentiated neurons than in neuroblastoma precursors suggests a differential neuronal vulnerability to heat. We characterized the neuronal heat shock response through RT-PCR array analysis of eighty-four genes involved in protein folding and protein quality control (PQC). We identify seventeen significantly expressed genes, five of which are Hsp70 chaperones, and four of their known complementing function proteins. Protein expression analysis determined the individual differential contribution of the five Hsp70 chaperones to the proteotoxic stress response and the significance of only two members under mild conditions. The co-expression analysis reveals significantly high co-expression between the Hsp70 chaperones and their interacting partners. The findings of this study lend support to the hypothesis that hyperthermia-induced proteotoxicity may underlie the brain injury of heat stroke. Additionally, this study presents a comprehensive map of the Hsp70 network in these models with potential clinical and translational implications.

6.
J Physiol ; 601(12): 2407-2423, 2023 06.
Article in English | MEDLINE | ID: mdl-36951421

ABSTRACT

An evolutionary heat shock response (HSR) protects most living species, including humans, from heat-induced macromolecular damage. However, its role in the pathogenesis of heat stroke is unknown. We examined the whole genome transcriptome in peripheral blood mononuclear cells of a cohort of subjects exposed to the same high environmental heat conditions, who developed heat stroke (n = 19) versus those who did not (n = 19). Patients with heat stroke had a mean rectal temperature at admission of 41.7 ± 0.8°C, and eight were in deep coma (Glasgow Coma Score = 3). The transcriptome showed that genes involved in more than half of the entire chaperome were differentially expressed relative to heat stress control. These include the heat shock protein, cochaperone, and chaperonin genes, indicating a robust HSR. Differentially expressed genes also encoded proteins related to unfolded protein response, DNA repair, energy metabolism, oxidative stress, and immunity. The analysis predicted perturbations of the proteome network and energy production. Cooling therapy attenuated these alterations without complete restoration of homeostasis. We validated the significantly expressed genes by a real-time polymerase chain reaction. The findings reveal the molecular signature of heat stroke. They also suggested that a powerful HSR may not be sufficient to protect against heat injury. The overwhelming proteotoxicity and energy failure could play a pathogenic role. KEY POINTS: Most living species, including humans, have inherent heat stress response (HSR) that shields them against heat-induced macromolecular damage. The role of the HSR in subjects exposed to environmental heat who progressed to heat stroke versus those that did not is unknown. Our findings suggest that heat stroke induces a broad and robust HSR of nearly half of the total heat shock proteins, cochaperones, and chaperonin genes. Heat stroke patients exhibited inhibition of genes involved in energy production, including oxidative phosphorylation and ATP production. Significant enrichment of neurodegenerative pathways, including amyloid processing signalling, the Huntington's and Parkinson's disease signalling suggestive of brain proteotoxicity was noted. The data suggests that more than a powerful HSR may be required to protect against heat stroke. Overwhelming proteotoxicity and energy failure might contribute to its pathogenesis.


Subject(s)
Heat Stroke , Transcriptome , Humans , Coma , Leukocytes, Mononuclear , Heat-Shock Response/genetics , Heat-Shock Proteins/genetics , Heat Stroke/genetics
7.
Cells ; 12(3)2023 01 19.
Article in English | MEDLINE | ID: mdl-36766718

ABSTRACT

G protein-coupled receptors (GPCRs) are expressed essentially on all cells, facilitating cellular responses to external stimuli, and are involved in nearly every biological process. Several members of this family play significant roles in the regulation of adipogenesis and adipose metabolism. However, the expression and functional significance of a vast number of GPCRs in adipose tissue are unknown. We used a high-throughput RT-PCR panel to determine the expression of the entire repertoire of non-sensory GPCRs in mouse white, and brown adipose tissue and assess changes in their expression during adipogenic differentiation of murine adipocyte cell line, 3T3-L1. In addition, the expression of GPCRs in subcutaneous adipose tissues from lean, obese, and diabetic human subjects and in adipocytes isolated from regular chow and high-fat fed mice were evaluated by re-analyzing RNA-sequencing data. We detected a total of 292 and 271 GPCRs in mouse white and brown adipose tissue, respectively. There is a significant overlap in the expression of GPCRs between the two adipose tissue depots, but several GPCRs are specifically expressed in one of the two tissue types. Adipogenic differentiation of 3T3-L1 cells had a profound impact on the expression of several GPCRs. RNA sequencing of subcutaneous adipose from healthy human subjects detected 255 GPCRs and obesity significantly changed the expression of several GPCRs in adipose tissue. High-fat diet had a significant impact on adipocyte GPCR expression that was similar to human obesity. Finally, we report several highly expressed GPCRs with no known role in adipose biology whose expression was significantly altered during adipogenic differentiation, and/or in the diseased human subjects. These GPCRs could play an important role in adipose metabolism and serve as a valuable translational resource for obesity and metabolic research.


Subject(s)
Adipocytes , Obesity , Humans , Mice , Animals , Adipocytes/metabolism , Obesity/metabolism , Cell Differentiation/genetics , Adipose Tissue, Brown/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
8.
J Endocr Soc ; 8(1): bvad159, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-38162016

ABSTRACT

Context: Bariatric surgery has been shown to be effective in inducing complete remission of type 2 diabetes in adults with obesity. However, its efficacy in achieving complete diabetes remission remains variable and difficult to predict before surgery. Objective: We aimed to characterize bariatric surgery-induced transcriptome changes associated with diabetes remission and the predictive role of the baseline transcriptome. Methods: We performed a whole-genome microarray in peripheral mononuclear cells at baseline (before surgery) and 2 and 12 months after bariatric surgery in a prospective cohort of 26 adults with obesity and type 2 diabetes. We applied machine learning to the baseline transcriptome to identify genes that predict metabolic outcomes. We validated the microarray expression profile using a real-time polymerase chain reaction. Results: Sixteen patients entered diabetes remission at 12 months and 10 did not. The gene-expression analysis showed similarities and differences between responders and nonresponders. The difference included the expression of critical genes (SKT4, SIRT1, and TNF superfamily), metabolic and signaling pathways (Hippo, Sirtuin, ARE-mediated messenger RNA degradation, MSP-RON, and Huntington), and predicted biological functions (ß-cell growth and proliferation, insulin and glucose metabolism, energy balance, inflammation, and neurodegeneration). Modeling the baseline transcriptome identified 10 genes that could hypothetically predict the metabolic outcome before bariatric surgery. Conclusion: The changes in the transcriptome after bariatric surgery distinguish patients in whom diabetes enters complete remission from those who do not. The baseline transcriptome can contribute to the prediction of bariatric surgery-induced diabetes remission preoperatively.

9.
Curr Issues Mol Biol ; 44(10): 4748-4768, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36286039

ABSTRACT

Placenta-derived stem cells (PDSCs), due to unique traits such as mesenchymal and embryonic characteristics and the absence of ethical constraints, are in a clinically and therapeutically advantageous position. To aid in stemness maintenance, counter pathophysiological stresses, and withstand post-differentiation challenges, stem cells require elevated protein synthesis and consequently augmented proteostasis. Stem cells exhibit source-specific proteostasis traits, making it imperative to study them individually from different sources. These studies have implications for understanding stem cell biology and exploitation in the augmentation of therapeutic applications. Here, we aim to identify the primary determinants of proteotoxic stress response in PDSCs. We generated heat-induced dose-responsive proteotoxic stress models of three stem cell types: placental origin cells, the placenta-derived mesenchymal stem cells (pMSCs), maternal origin cells, the decidua parietalis mesenchymal stem cells (DPMSCs), and the maternal-fetal interface cells, decidua basalis mesenchymal stem cells (DBMSCs), and measured stress induction through biochemical and cell proliferation assays. RT-PCR array analysis of 84 genes involved in protein folding and protein quality control led to the identification of Hsp70 members HSPA1A and HSPA1B as the prominent ones among 17 significantly expressed genes and with further analysis at the protein level through Western blotting. A kinetic analysis of HSPA1A and HSPA1B gene and protein expression allowed a time series evaluation of stress response. As identified by protein expression, an active stress response is in play even at 24 h. More prominent differences in expression between the two homologs are detected at the translational level, alluding to a potential higher requirement for HSPA1B during proteotoxic stress response in PDSCs.

10.
Micromachines (Basel) ; 13(9)2022 Sep 03.
Article in English | MEDLINE | ID: mdl-36144084

ABSTRACT

Lychee (Litchi chinensis Sonn.) is a famous fruit species of tropical and subtropical regions of the world and many biotic and abiotic stresses affect its yield. In this study, lychee fruit rot has been observed and its incidence has been controlled by using zinc oxide nanoparticles (ZnO NPs). Diseased lychee fruits were collected and diagnosed to identify disease-causing pathogens. Morphological appearance, microscopic observation, and sequence analysis of the amplified ITS region identified this isolated pathogen as Aspergillus niger. To control this problem, ZnO NPs were prepared in the leaf extract of Azadirachta indica. Before their antifungal activity, ZnO NPs were characterized using sophisticated approaches. FTIR revealed the presence of reducing and stabilizing molecules on ZnO NPs including alcohol, carboxylic acid, alkyl halide, amine, and alkyl halide. Crystalline nature and average size (29.024 nm) of synthesized ZnO NPs were described by X-ray diffraction. EDX analysis depicted the mass percentage of zinc (30.15%) and oxygen (14.90%). SEM analysis displayed the irregular shape of nanoparticles and confirmed the nano-size of ZnO NPs. Maximum mycelial growth inhibition (70.5%) was observed at 1.0 mg/mL concentration of ZnO NPs in vitro. In in-vivo disease-control analysis, maximum control of lychee fruit rot disease was observed at the same concentration. These results reveal the potential use of these ZnO NPs on a larger scale to replace hazardous chemical fungicides.

11.
Molecules ; 27(14)2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35889333

ABSTRACT

Cherry is a fleshy drupe, and it is grown in temperate regions of the world. It is perishable, and several biotic and abiotic factors affect its yield. During April-May 2021, a severe fruit rot of cherry was observed in Swat and adjacent areas. Diseased fruit samples were collected, and the disease-causing pathogen was isolated on PDA. Subsequent morphological, microscopic, and molecular analyses identified the isolated pathogen as Aspergillus flavus. For the control of the fruit rot disease of cherry, iron oxide nanoparticles (Fe2O3 NPs) were synthesized in the leaf extract of Calotropis procera and characterized. Fourier transform infrared (FTIR) spectroscopy of synthesized Fe2O3 NPs showed the presence of capping and stabilizing agents such as alcohols, aldehydes, and halo compounds. X-ray diffraction (XRD) analysis verified the form and size (32 nm) of Fe2O3 NPs. Scanning electron microscopy (SEM) revealed the spinal-shaped morphology of synthesized Fe2O3 NPs while X-ray diffraction (EDX) analysis displayed the occurrence of main elements in the samples. After successful preparation and characterization of NPs, their antifungal activity against A. flavus was determined by poison technique. Based on in vitro and in vivo antifungal activity analyses, it was observed that 1.0 mg/mL concentration of Fe2O3 can effectively inhibit the growth of fungal mycelia and decrease the incidence of fruit rot of cherry. The results confirmed ecofriendly fungicidal role of Fe2O3 and suggested that their large-scale application in the field to replace toxic chemical fungicides.


Subject(s)
Calotropis , Metal Nanoparticles , Nanoparticles , Anti-Bacterial Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/pharmacology , Fruit , Metal Nanoparticles/chemistry , Nanoparticles/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
12.
Curr Cancer Drug Targets ; 22(7): 577-590, 2022.
Article in English | MEDLINE | ID: mdl-35585825

ABSTRACT

AIM: The study aims to understand the role of tumor suppressor genes in colorectal cancer initiation and progression. BACKGROUND: Sporadic colorectal cancer (CRC) develops through distinct molecular events. Loss of the 18q chromosome is a conspicuous event in the progression of adenoma to carcinoma. There is limited information regarding the molecular effectors of this event. Earlier, we had reported ATP8B1 as a novel gene associated with CRC. ATP8B1 belongs to the family of P-type ATPases (P4 ATPase) that primarily function to facilitate the translocation of phospholipids. OBJECTIVE: In this study, we attempt to implicate the ATP8B1 gene located on chromosome 18q as a tumor suppressor gene. METHODS: Cells culture, Patient data analysis, Generation of stable ATP8B1 overexpressing SW480 cell line, Preparation of viral particles, Cell Transduction, Generation of stable ATP8B1 knockdown HT29 cell line with CRISPR/Cas9, Generation of stable ATP8B1 knockdown HT29 cell line with shRNA, Quantification of ATP8B1 gene expression, Real-time cell proliferation and migration assays, Cell proliferation assay, Cell migration assay, Protein isolation and western blotting, Endpoint cell viability assay, Uptake and efflux of sphingolipid, Statistical and computational analyses. RESULTS: We studied indigenous patient data and confirmed the reduced expression of ATP8B1 in tumor samples. CRC cell lines were engineered with reduced and enhanced levels of ATP8B1, which provided a tool to study its role in cancer progression. Forced reduction of ATP8B1 expression either by CRISPR/Cas9 or shRNA was associated with increased growth and proliferation of CRC cell line - HT29. In contrast, overexpression of ATP8B1 resulted in reduced growth and proliferation of SW480 cell lines. We generated a network of genes that are downstream of ATP8B1. Further, we provide the predicted effect of modulation of ATP8B1 levels on this network and the possible effect on fatty acid metabolism-related genes. CONCLUSION: Tumor suppressor gene (ATP8B1) located on chromosome 18q could be responsible in the progression of colorectal cancer. Knocking down of this gene causes an increased rate of cell proliferation and reduced cell death, suggesting its role as a tumor suppressor. Increasing the expression of this gene in colorectal cancer cells slowed down their growth and increased cell death. These evidences suggest the role of ATP8B1 as a tumor suppressor gene.


Subject(s)
Colorectal Neoplasms , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , RNA, Small Interfering/genetics
13.
Cells ; 11(4)2022 02 21.
Article in English | MEDLINE | ID: mdl-35203397

ABSTRACT

Fatty acids (FFAs) are important biological molecules that serve as a major energy source and are key components of biological membranes. In addition, FFAs play important roles in metabolic regulation and contribute to the development and progression of metabolic disorders like diabetes. Recent studies have shown that FFAs can act as important ligands of G-protein-coupled receptors (GPCRs) on the surface of cells and impact key physiological processes. Free fatty acid-activated receptors include FFAR1 (GPR40), FFAR2 (GPR43), FFAR3 (GPR41), and FFAR4 (GPR120). FFAR2 and FFAR3 are activated by short-chain fatty acids like acetate, propionate, and butyrate, whereas FFAR1 and FFAR4 are activated by medium- and long-chain fatty acids like palmitate, oleate, linoleate, and others. FFARs have attracted considerable attention over the last few years and have become attractive pharmacological targets in the treatment of type 2 diabetes and metabolic syndrome. Several lines of evidence point to their importance in the regulation of whole-body metabolic homeostasis including adipose metabolism. Here, we summarize our current understanding of the physiological functions of FFAR isoforms in adipose biology and explore the prospect of FFAR-based therapies to treat patients with obesity and Type 2 diabetes.


Subject(s)
Adipose Tissue , Diabetes Mellitus, Type 2 , Fatty Acids, Nonesterified , Receptors, G-Protein-Coupled , Adipose Tissue/metabolism , Fatty Acids, Nonesterified/metabolism , Humans , Ligands , Obesity , Receptors, G-Protein-Coupled/metabolism
14.
Pharmaceutics ; 15(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36678745

ABSTRACT

After the successful commercial exploitation of 3D printing technology, the advanced version of additive manufacturing, i.e., 4D printing, has been a new buzz in the technology-driven industries since 2013. It is a judicious combination of 3D printing technologies and smart materials (stimuli responsive), where time is the fourth dimension. Materials such as liquid crystal elastomer (LCE), shape memory polymers, alloys and composites exhibiting properties such as self-assembling and self-healing are used in the development/manufacturing of these products, which respond to external stimuli such as solvent, temperature, light, etc. The technologies being used are direct ink writing (DIW), fused filament fabrication (FFF), etc. It offers several advantages over 3D printing and has been exploited in different sectors such as healthcare, textiles, etc. Some remarkable applications of 4D printing technology in healthcare are self-adjusting stents, artificial muscle and drug delivery applications. Potential of applications call for further research into more responsive materials and technologies in this field. The given review is an attempt to collate all the information pertaining to techniques employed, raw materials, applications, clinical trials, recent patents and publications specific to healthcare products. The technology has also been evaluated in terms of regulatory perspectives. The data garnered is expected to make a strong contribution to the field of technology for human welfare and healthcare.

16.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34072007

ABSTRACT

Adrenergic receptor ß3 (ADRß3) is a member of the rhodopsin-like G protein-coupled receptor family. The binding of the ligand to ADRß3 activates adenylate cyclase and increases cAMP in the cells. ADRß3 is highly expressed in white and brown adipocytes and controls key regulatory pathways of lipid metabolism. Trp64Arg (W64R) polymorphism in the ADRß3 is associated with the early development of type 2 diabetes mellitus, lower resting metabolic rate, abdominal obesity, and insulin resistance. It is unclear how the substitution of W64R affects the functioning of ADRß3. This study was initiated to functionally characterize this obesity-linked variant of ADRß3. We evaluated in detail the expression, subcellular distribution, and post-activation behavior of the WT and W64R ADRß3 using single cell quantitative fluorescence microscopy. When expressed in HEK 293 cells, ADRß3 shows a typical distribution displayed by other GPCRs with a predominant localization at the cell surface. Unlike adrenergic receptor ß2 (ADRß2), agonist-induced desensitization of ADRß3 does not involve loss of cell surface expression. WT and W64R variant of ADRß3 displayed comparable biochemical properties, and there was no significant impact of the substitution of tryptophan with arginine on the expression, cellular distribution, signaling, and post-activation behavior of ADRß3. The obesity-linked W64R variant of ADRß3 is indistinguishable from the WT ADRß3 in terms of expression, cellular distribution, signaling, and post-activation behavior.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Obesity/genetics , Obesity/metabolism , Receptors, Adrenergic, beta-3/genetics , Receptors, Adrenergic, beta-3/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Alleles , Amino Acid Substitution , Cell Line , Gene Expression , Genetic Association Studies , Humans , Mutation , Polymorphism, Genetic , Protein Transport
17.
Immun Ageing ; 18(1): 1, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33390183

ABSTRACT

The disease (COVID-19) novel coronavirus pandemic has so far infected millions resulting in the death of over a million people as of Oct 2020. More than 90% of those infected with COVID-19 show mild or no symptoms but the rest of the infected cases show severe symptoms resulting in significant mortality. Age has emerged as a major factor to predict the severity of the disease and mortality rates are significantly higher in elderly patients. Besides, patients with underlying conditions like Type 2 diabetes, cardiovascular diseases, hypertension, and cancer have an increased risk of severe disease and death due to COVID-19 infection. Obesity has emerged as a novel risk factor for hospitalization and death due to COVID-19. Several independent studies have observed that people with obesity are at a greater risk of severe disease and death due to COVID-19. Here we review the published data related to obesity and overweight to assess the possible risk and outcome in Covid-19 patients based on their body weight. Besides, we explore how the obese host provides a unique microenvironment for disease pathogenesis, resulting in increased severity of the disease and poor outcome.

18.
Pathogens ; 9(9)2020 Aug 20.
Article in English | MEDLINE | ID: mdl-32825438

ABSTRACT

The COVID-19 pandemic, in the first seven months, has led to more than 15 million confirmed infected cases and 600,000 deaths. SARS-CoV-2, the causative agent for COVID-19, has proved to be a great challenge for its ability to spread in asymptomatic stages and the diverse disease spectrum it has generated. This has created a challenge of unimaginable magnitude, not only affecting human health and life but also potentially generating a long-lasting socioeconomic impact. Both medical sciences and biomedical research have also been challenged, consequently leading to a large number of clinical trials and vaccine initiatives. While known proteins of pathobiological importance are targets for these therapeutic approaches, it is imperative to explore other factors of viral significance. Accessory proteins are one such trait that have diverse roles in coronavirus pathobiology. Here, we analyze certain genomic characteristics of SARS-CoV-2 accessory protein ORF8 and predict its protein features. We have further reviewed current available literature regarding its function and comparatively evaluated these and other features of ORF8 and ORF8ab, its homolog from SARS-CoV. Because coronaviruses have been infecting humans repeatedly and might continue to do so, we therefore expect this study to aid in the development of holistic understanding of these proteins. Despite low nucleotide and protein identity and differentiating genome level characteristics, there appears to be significant structural integrity and functional proximity between these proteins pointing towards their high significance. There is further need for comprehensive genomics and structural-functional studies to lead towards definitive conclusions regarding their criticality and that can eventually define their relevance to therapeutics development.

19.
World J Gastrointest Oncol ; 12(5): 514-525, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32461783

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a worldwide problem, which has been associated with changes in diet and lifestyle pattern. As a result of colonic fermentation of dietary fibres, short chain free fatty acids are generated which activate free fatty acid receptors (FFAR) 2 and 3. FFAR2 and FFAR3 genes are abundantly expressed in colonic epithelium and play an important role in the metabolic homeostasis of colonic epithelial cells. Earlier studies point to the involvement of FFAR2 in colorectal carcinogenesis. AIM: To understand the role of short chain FFARs in CRC. METHODS: Transcriptome analysis console software was used to analyse microarray data from CRC patients and cell lines. We employed short-hairpin RNA mediated down regulation of FFAR2 and FFAR3 genes, which was validated using quantitative real time polymerase chain reaction. Assays for glucose uptake and cyclic adenosine monophosphate (cAMP) generation was done along with immunofluorescence studies to study the effects of FFAR2/FFAR3 knockdown. For measuring cell proliferation, we employed real time electrical impedance-based assay available from xCELLigence. RESULTS: Microarray data analysis of CRC patient samples showed a significant down regulation of FFAR2 gene expression. This prompted us to study the FFAR2 in CRC. Since, FFAR3 shares significant structural and functional homology with FFAR2, we knocked down both these receptors in CRC cell line HCT 116. These modified cell lines exhibited higher proliferation rate and were found to have increased glucose uptake as well as increased level of glucose transporter 1. Since, FFAR2 and FFAR3 signal through G protein subunit (Gαi), knockdown of these receptors was associated with increased cAMP. Inhibition of protein kinase A (PKA) did not alter the growth and proliferation of these cells indicating a mechanism independent of cAMP/PKA pathway. CONCLUSION: Our results suggest role of FFAR2/FFAR3 genes in increased proliferation of colon cancer cells via enhanced glucose uptake and exclude the role of PKA mediated cAMP signalling. Alternate pathways could be involved that would ultimately result in increased cell proliferation as a result of down regulated FFAR2/FFAR3 genes. This study paves the way to understand the mechanism of action of short chain FFARs in CRC.

20.
Curr Mol Med ; 20(6): 488-493, 2020.
Article in English | MEDLINE | ID: mdl-31840614

ABSTRACT

BACKGROUND: G protein-coupled receptors (GPCRs) represent the largest family of surface proteins and are involved in the regulation of key physiological processes. GPCRs are characterized by seven transmembrane domains, an extracellular N-terminus and an intracellular C-terminus. Cellular response of these receptors to their ligands is largely determined by their surface expression and postactivation behavior including expression, desensitization and resensitization. OBJECTIVE: To develop a quantitative fluorescence Microscopy assay to study ß2- Adrenergic receptor expression and desensitization. METHOD: ß2-Adrenergic receptor cDNA was engineered to put an HA tag at the extracellular N-terminus and GFP Tag at the intracellular C-terminus. GFP fluorescence serves as a measure of total cellular expression; whereas staining with CY3 conjugated anti-HA antibodies without permeabilizing the cells represents the surface expression of ß2-AR. The images are quantified and amount of CY3 (surface) and GFP (total) fluorescence for each cell determined using image processing software. RESULTS: The method is sensitive and allows for the simultaneous measurement of surface and total expression of ß2-AR. CONCLUSION: A highly accurate method is described for measuring ß2-AR surface and total expression based on single-cell quantitative immunofluorescence. The method can be used to determine agonist-induced desensitization and resensitization process as well as receptor kinetics like endocytosis and exocytosis of ß2-Adrenergic receptor and can be applied to essentially any other GPCR.


Subject(s)
Receptors, Adrenergic, beta-2/metabolism , Animals , Humans , Microscopy, Fluorescence , Receptors, Adrenergic, beta-2/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Single-Cell Analysis , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...