SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Tripathi Rekha PhD student 1985 ) "

Sökning: WFRF:(Tripathi Rekha PhD student 1985 )

  • Resultat 1-7 av 7
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Schuster, Jens, Assistant Professor, 1972-, et al. (författare)
  • Generation of a human iPSC line (UUIGPi015-A) from a patient with Dravet syndrome and a 2.9 Mb deletion spanning SCN1A on chromosome 2
  • 2022
  • Ingår i: Stem Cell Research. - : Elsevier. - 1873-5061 .- 1876-7753. ; 60
  • Tidskriftsartikel (refereegranskat)abstract
    • Dravet syndrome is an early onset devastating epilepsy syndrome usually caused by heterozygous mutations in SCN1A. We generated a human iPSC line (UUIGPi015A) from dermal fibroblasts of a patient with Dravet syndrome carrying a deletion on chromosome 2 encompassing SCN1A and 9 flanking genes. Characterization of the iPSC line confirmed expression of pluripotency markers, tri-lineage differentiation capacity and absence of exogenous reprogramming factors. The iPSC line retained the deletion and was genomically stable. The iPSC line UUIGPi015-A provides a useful resource for studies on the pathophysiology of Dravet syndrome and seizures caused by haploinsufficiency of SCN1A and flanking gene products.
  •  
2.
  • Han, Yilin, et al. (författare)
  • Molecular genetic analysis of neural stem cells after space flight and simulated microgravity on earth
  • 2021
  • Ingår i: Biotechnology and Bioengineering. - : John Wiley & Sons. - 0006-3592 .- 1097-0290. ; 118:10, s. 3832-3846
  • Tidskriftsartikel (refereegranskat)abstract
    • Understanding how stem cells adapt to space flight conditions is fundamental for human space missions and extraterrestrial settlement. We analyzed gene expression in boundary cap neural crest stem cells (BCs), which are attractive for regenerative medicine by their ability to promote proliferation and survival of cocultured and co-implanted cells. BCs were launched to space (space exposed cells) (SEC), onboard sounding rocket MASER 14 as free-floating neurospheres or in a bioprinted scaffold. For comparison, BCs were placed in a random positioning machine (RPM) to simulate microgravity on earth (RPM cells) or were cultured under control conditions in the laboratory. Using next-generation RNA sequencing and data post-processing, we discovered that SEC upregulated genes related to proliferation and survival, whereas RPM cells upregulated genes associated with differentiation and inflammation. Thus, (i) space flight provides unique conditions with distinctly different effects on the properties of BC compared to earth controls, and (ii) the space flight exposure induces postflight properties that reinforce the utility of BC for regenerative medicine and tissue engineering.
  •  
3.
  • Tripathi, Rekha, PhD student, 1985-, et al. (författare)
  • Integrated analysis of transcriptomic and metabolic profiling of SLC38A10 knockout  primary cortex cells
  • Annan publikation (övrigt vetenskapligt/konstnärligt)abstract
    • Solute carrier transporter responsible protein to transfer of biological membrane and act as a vital regulator of transporter of amino acid, sugar, ion, and drug. Members of the glutamine transporters SLC38 family have played a key role in regulating the Glutamate/GABA-glutamine cycle (GGG) and nutrient-sensing mTOR pathway. Membrane-bound SLC38A10 transporters regulate the flow of glutamine, glutamate, aspartic acid, and alanine and play a vital role in regulating neurotransmission. This study aimed to understand the possible role of SLC38A10 in brain metabolism and nutrient-sensing pathway. We have used primary cortex cultured from SLC38 Knockout mice and performed nutrient starvation and refed experiment. Here we used the omics approach to integrate data derived from transcriptomic and metabolomics. Differentially expressed genes and changed metabolites were submitted in ingenuity pathway analysis (IPA) to elucidate the cellular function and disease resulting from the absence of SLC38A10 in primary cortex cells.Further, the effect of nutrient deprivation on KO PCCs cells was studied, and associated cellular pathway and bio function indicated towards disease. Our findings relevel that KO cells increase neurotransmission, GABA receptor signalling, Synaptic transmission, Neurotransmission of cerebral cortex cells, Differentiation of oligodendrocytes, neuroglia, branching of neurons, apoptosis and degeneration of neurons, Sprouting, Branching of neurites, dendritic growth/branching and Concentration of lipid. The overall conclusion suggests that SLC38A10 removal results in damaged neuronal morphology and nervous system morphology and functionality, linked with neurodevelopmental and neurological disease.
  •  
4.
  • Tripathi, Rekha, PhD student, 1985-, et al. (författare)
  • SLC38A10 Regulate Glutamate Homeostasis and Modulate the AKT/TSC2/mTOR Pathway in Mouse Primary Cortex Cells
  • 2022
  • Ingår i: Frontiers in cell and developmental biology. - : Frontiers Media S.A.. - 2296-634X. ; 10
  • Tidskriftsartikel (refereegranskat)abstract
    • Glutamate acts as a critical regulator of neurotransmitter balance, recycling, synaptic function and homeostasis in the brain and glutamate transporters control glutamate levels in the brain. SLC38A10 is a member of the SLC38 family and regulates protein synthesis and cellular stress responses. Here, we uncover the role of SLC38A10 as a transceptor involved in glutamate-sensing signaling pathways that control both the glutamate homeostasis and mTOR-signaling. The culture of primary cortex cells from SLC38A10 knockout mice had increased intracellular glutamate. In addition, under nutrient starvation, KO cells had an impaired response in amino acid-dependent mTORC1 signaling. Combined studies from transcriptomics, protein arrays and metabolomics established that SLC38A10 is involved in mTOR signaling and that SLC38A10 deficient primary cortex cells have increased protein synthesis. Metabolomic data showed decreased cholesterol levels, changed fatty acid synthesis, and altered levels of fumaric acid, citrate, 2-oxoglutarate and succinate in the TCA cycle. These data suggests that SLC38A10 may act as a modulator of glutamate homeostasis, and mTOR-sensing and loss of this transceptor result in lower cholesterol, which could have implications in neurodegenerative diseases.
  •  
5.
  • Tripathi, Rekha, PhD student, 1985-, et al. (författare)
  • SLC38A10 Transporter Plays a Role in Cell Survival Under Oxidative Stress and Glutamate Toxicity
  • 2021
  • Ingår i: Frontiers in Bioscience. - : Frontiers Media S.A.. - 1093-9946 .- 1093-4715 .- 2296-889X. ; 8
  • Tidskriftsartikel (refereegranskat)abstract
    • Solute carrier (SLC) transporters regulate amino acids, glucose, ions, and metabolites that flow across cell membranes. In the brain, SLCs are the key regulators of neurotransmission, in particular, the glutamate/GABA-glutamine (GGG) cycle. Genetic mutations in SLCs are associated with various neurodevelopmental and neurodegenerative diseases. In this study, we have investigated the role of SLC38A10 under acute oxidative and glutamate stress in mouse primary cortical cells from SLC38A10 knockout (KO) mice. The ER/golgi localized transporter, SLC38A10, transports glutamate, glutamine, and alanine in brain cells, and the aim of this study was to determine the possible effects of removal of SLC38A10 in primary cortical cells under glutamate and oxidative challenges. Primary cortical neuronal cultures of wild-type (WT) cell and SLC38A10 KO mice were subjected to different concentrations of glutamate and hydrogen peroxide. There was no morphological change observed between KO and WT cortical neurons in culture. Interestingly, KO cells showed significantly lower cell viability and higher cell death compared to WT cells under both glutamate and hydrogen peroxide exposure. Further, we evaluated the possible role of p53 in neuronal cell apoptosis in KO cells. We found decreased intracellular p53 protein levels under glutamate and hydrogen peroxide treatment in KO cortical cells. In contrast, caspase 3/7 activity remains unaltered under all conditions. These results demonstrate an indirect relationship between the expression of SLC38A10 and p53 and a role in the cell defense mechanism against neurotoxicity.
  •  
6.
  •  
7.
  • Tripathi, Rekha, PhD student, 1985- (författare)
  • Unlocking the Role Of Orphan Solute Carrier SLC38A10 In Brain Metabolism : The SLC38A10 transporter in nutrient and metabolic regulation
  • 2021
  • Doktorsavhandling (övrigt vetenskapligt/konstnärligt)abstract
    • Membrane transporters are the primary gatekeepers of cells and regulate the transport of nutrients, metabolites, ions, water, and neurotransmitters into and across the human cells. The solute carrier transporters (SLCs) are the most prominent transporters, comprising 430 members divided into 65 subfamilies. SLCs are located on the plasma membrane and organelles such as mitochondria, vesicles, peroxisomes, endoplasmic reticulum, Golgi, and lysosomes. This thesis aimed to study SLCs of the SLC38 family under nutrient stress, focused particularly on the orphan SLC38A10 transporter.In Paper I, regulation of members of SLC38 family transporter, after amino acid starvation in mouse hypothalamic cells and primary cortex cells, was studied using microarrays and qPCR. We found several members of the SLC38 family that were strongly affected under amino acid starvation and showing a potential role in amino acid signaling in the brain. In Paper II, we performed a cellular and tissue localization and functional study of SLC38A10 transporter and revealed that SLC38A10 was expressed in both excitatory and inhibitory neurons in the mouse brain and has a unique subcellular localization in the ER and Golgi membrane. Furthermore, knockdown of the SLC38A10 gene resulted in reduced nascent protein synthesis in PC12 cells. Further, to unlock the biological function of the SLC38A10 transporter, in Paper III and Paper IV, we used SLC38A10 knockout mouse model.In Paper III, the goal was to uncover the role of SLC38A10 in acute glutamate and oxidative stress. Here, we found that a loss of SLC38A10 KO resulted in changes in the p53 levels and affected the mitochondrial function. Thus, this study established a possible role of SLC38A10 in cell survival, linked with p53, in mouse primary cortex cells. In Paper IV, we examined the role of SLC38A10 in amino acid metabolism and nutrient sensing in the mTOR signaling pathway. We performed complete amino acid starvation and refeed experiment on SLC38A10 knockout primary cortex cells. We concluded that SLC38A10 acts as a transceptor and regulates mTOR-dependent protein and lipid synthesis in brain cells, corroborating the findings from Paper II. To summarize, the present work has uncovered the function of SLC38A10 in the brain. It also provides knowledge of SLC38A10’s role in amino acid metabolism and signaling pathway(s). The findings of this thesis will enhance an understanding of SLC38A10 transporter and provide insight into future disease targeted drug studies focused on metabolic disorder and neurodegenerative disease.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-7 av 7

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy