Summary

Measuring Endoplasmic Reticulum Stress and Unfolded Protein Response in HIV-1 Infected T-Cells and Analyzing its Role in HIV-1 Replication

Published: June 14, 2024
doi:

Summary

Here, we describe some established methods to determine endoplasmic reticulum (ER) stress and unfolded protein response (UPR) activation, with particular emphasis on HIV-1 infection. This article also describes a set of protocols to investigate the effect of ER stress/UPR on HIV-1 replication and virion infectivity.

Abstract

Viral infections can cause Endoplasmic Reticulum (ER) stress due to abnormal protein accumulation, leading to Unfolded Protein Response (UPR). Viruses have developed strategies to manipulate the host UPR, but there is a lack of detailed understanding of UPR modulation and its functional significance during HIV-1 infection in the literature. In this context, the current article describes the protocols used in our laboratory to measure ER stress levels and UPR during HIV-1 infection in T-cells and the effect of UPR on viral replication and infectivity.

Thioflavin T (ThT) staining is a relatively new method used to detect ER stress in the cells by detecting protein aggregates. Here, we have illustrated the protocol for ThT staining in HIV-1 infected cells to detect and quantify ER stress. Moreover, ER stress was also detected indirectly by measuring the levels of UPR markers such as BiP, phosphorylated IRE1, PERK, and eIF2α, splicing of XBP1, cleavage of ATF6, ATF4, CHOP, and GADD34 in HIV-1 infected cells, using conventional immunoblotting and quantitative reverse transcription polymerase chain reaction (RT-PCR). We have found that the ThT-fluorescence correlates with the indicators of UPR activation. This article also demonstrates the protocols to analyze the impact of ER stress and UPR modulation on HIV-1 replication by knockdown experiments as well as the use of pharmacological molecules. The effect of UPR on HIV-1 gene expression/replication and virus production was analyzed by Luciferase reporter assays and p24 antigen capture ELISA, respectively, whereas the effect on virion infectivity was analyzed by staining of infected reporter cells. Collectively, this set of methods provides a comprehensive understanding of the Unfolded Protein Response pathways during HIV-1 infection, revealing its intricate dynamics.

Introduction

Acquired immunodeficiency syndrome (AIDS) is characterized by a gradual reduction in the number of CD4+ T-lymphocytes, which leads to the progressive failure of immune response. Human immunodeficiency virus-1 (HIV-1) is the causative agent of AIDS. It is an enveloped, positive sense, single-stranded RNA virus with two copies of RNA per virion and belongs to the retroviridae family. Production of high concentrations of viral proteins within the host cell places excessive stress on the protein folding machinery of the cell1. ER is the first compartment in the secretory pathway of eukaryotic cells. It is in charge of producing, altering, and delivering proteins to the secretory pathway and the extracellular space target sites. Proteins undergo numerous post-translational changes and fold into their natural conformation in the ER, including asparagine-linked glycosylation and the creation of intra- and intermolecular disulfide bonds2. Therefore, high concentrations of proteins are present in the ER lumen and these are very prone to aggregation and misfolding. Various physiological conditions, such as heat shock, microbial, or viral infections, which demand enhanced protein synthesis or protein mutation, lead to ER stress due to increased protein accumulation in the ER, thereby disturbing the ER lumen homeostasis. The ER stress activates a network of highly conserved adaptive signal transduction pathways, the Unfolded Protein Response (UPR)3. UPR is employed to bring back the normal ER physiological condition by aligning its unfolded protein burden and folding capacity. This is brought upon by increasing the ER size and ER-resident molecular chaperones and foldases, resulting in an elevation of the ER's folding ability. UPR also decreases the protein load of the ER through global protein synthesis attenuation at the translational level and increases clearance of unfolded proteins from the ER by upregulating ER-associated degradation (ERAD)4,5.

ER stress is sensed by three ER-resident transmembrane proteins: Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), Activating transcription factor 6 (ATF6), and Inositol-requiring enzyme type 1 (IRE1). All these effectors are kept inactive by binding to the chaperone Heat shock protein family A (Hsp70) member 5 (HSPA5), also known as binding protein (BiP)/78-kDa glucose-regulated protein (GRP78). Upon ER stress and accumulation of unfolded/misfolded proteins, HSPA5 dissociates and leads to activation of these effectors, which then activate a series of downstream targets that help in resolving the ER stress and, in extreme conditions, promote cell death6. Upon dissociation from HSPA5, PERK autophosphorylates, and its kinase activity is activated7. Its kinase activity phosphorylates eIF2α, which leads to translational attenuation, lowering the protein load of the ER8. However, in the presence of phospho-eukaryotic initiation factor 2α (eIF2α), non-translated open reading frames on specific mRNAs become preferentially translated, such as ATF4, regulating stress-induced genes. ATF4 and C/EBP homologous protein (CHOP) are transcription factors that regulate stress-induced genes and regulate apoptosis and cell death pathways9,10. One of the targets of ATF4 and CHOP is the growth arrest and DNA damage-inducible protein (GADD34), which, along with protein phosphatase 1 dephosphorylate peIF2α and acts as a feedback regulator for translational attenuation11. Under ER stress, ATF6 dissociates from HSPA5, and its Golgi-localization signal is exposed, leading to its translocation to Golgi apparatus. In the Golgi apparatus, ATF6 is cleaved by site-1 protease (S1P) and site-2 protease (S2P) to release the cleaved form of ATF6 (ATF6 P50). ATF6 p50 is then translocated to the nucleus, where it induces the expression of genes involved in protein folding, maturation, and secretion as well as protein degradation12,13. During ER stress, IRE1 dissociates from HSPA5, multimerizes, and auto-phosphorylates14. Phosphorylation of IRE1 activates its RNase domain, specifically mediating the splicing of 26 nucleotides from the central part of the mRNA of X-box binding protein 1 (XBP1)15,16. This generates a novel C-terminus conferring transactivation function, generating functional XBP1s protein, a potent transcription factor controlling several ER stress-induced genes17,18. The combined activity of these transcription factors switches on genetic programs aimed at restoring ER homeostasis.

There are various methods to detect ER stress and UPR. These include the conventional methods of analyzing the UPR markers19,20. Various non-conventional methods include measuring the redox state of the UPR and calcium distribution in the ER lumen as well as assessing the ER structure. Electron microscopy may be used to see how much the ER lumen enlarges in response to ER stress in cells and tissues. However, this method is time-consuming and depends on the availability of an electron microscope, which may not be available to every research group. Also, measuring the calcium flux and the redox state of the ER is challenging due to the availability of reagents. Moreover, the readout from these experiments is very sensitive and might be affected by other factors of cellular metabolism.

A powerful and simple technique for monitoring the UPR outputs is to measure the activation of the different signaling pathways of the UPR and has been used for decades in various stress scenarios. These conventional methods to measure UPR activation are economical, feasible, and provide the information in less time as compared to other known methods. These include immunoblotting to measure the expression of UPR markers at the protein level, such as phosphorylation of IRE1, PERK, and eIF2α and cleavage of ATF6 by measuring the P50 form of ATF6 and protein expression of other markers such as HSPA5, spliced XBP1, ATF4, CHOP and GADD34 as well as RT-PCR to determine the mRNA levels as well as splicing of XBP1 mRNA.

This article describes a validated and reliable set of protocols to monitor ER stress and UPR activation in HIV-1 infected cells and to determine the functional relevance of UPR in HIV-1 replication and infectivity. The protocols utilize easily available as well as economical reagents and provide convincing information about the UPR outputs. ER stress is the result of the accumulation of unfolded/misfolded proteins, which are prone to forming protein aggregates21. We hereby describe a method to detect these protein aggregates in HIV-1-infected cells. Thioflavin T staining is a relatively new method being used to detect and quantify these protein aggregates22. Beriault and Werstuck described this technique to detect and quantify protein aggregates and, hence ER stress levels in live cells. It has been demonstrated that the small fluorescent molecule thioflavin T (ThT) binds selectively to protein aggregates, especially amyloid fibrils.

In this article, we describe the use of ThT to detect and quantify ER stress in HIV-1 infected cells and correlate it to the conventional method of monitoring UPR by measuring the activation of different signaling pathways of UPR.

Since, there is also a lack of comprehensive information regarding the role of UPR during HIV-1 infection, we provide a set of protocols to understand the role of UPR in HIV-1 replication and virion infectivity. These protocols include the lentivirus mediated knockdown of UPR markers as well as treatment with pharmacological ER stress inducers. This article also shows the types of read-out which can be used to measure the HIV-1 gene expression, viral production as well as the infectivity of the produced virions, such as long terminal repeat (LTR)-based luciferase assay, p24 enzyme-linked immunosorbent assay (ELISA) and β-gal reporter staining assay respectively.

Using the majority of these protocols, we have recently reported the functional implication of HIV-1 infection on UPR in T-cells23, and the results of that article suggest the reliability of the methods described here. Thus, this article provides a set of methods for comprehensive information regarding the interplay of HIV-1 with ER stress and UPR activation.

Protocol

NOTE: The cell lines used here are HEK-293T and Jurkat J6 (a CD4+T cell line), which were obtained from the Cell Repository, NCCS, Pune, India; TZM-bl, a HeLa derived cell line that has integrated copies of β-galactosidase and luciferase genes under the HIV-1 long terminal repeat (LTR) promoter24 and CEM-GFP (another CD4+ T reporter cell line)25 were obtained from the NIH AIDS Repository, USA. 1. HIV-1 virus stock preparation and stora…

Representative Results

In this work, we have described a detailed protocol to study in vitro ER stress and UPR activation upon HIV-1 infection in T-cells (Figure 2). This study also describes methods to analyze the functional relevance of UPR in HIV-1 replication and virion infectivity (Figure 3). To this purpose, we analyzed the ER stress caused by HIV-1 infection by observing the protein aggregates inside the cell by staining with Thioflavin T. A…

Discussion

The scope of the present protocol includes (i) the handling of HIV-1 virus stocks and the measurement of the virus concentration and virion infectivity, (ii) Infection of T-cells with HIV-1 and assessing its effect on ER stress and different markers of UPR, (iii) Effect of knockdown of UPR markers and their effect on HIV-1 LTR driven gene activity, virus production and virion infectivity and (iv) Overstimulating the UPR using pharmacological molecule and analyzing its effect on HIV-1 replication. Using the present set of…

Divulgations

The authors have nothing to disclose.

Acknowledgements

We thank the National Centre for Cell Science, Department of Biotechnology, Government of India, for intramural support. AT and AD are grateful for the Ph.D. research support received from the National Centre for Cell Science, Department of Biotechnology, Government of India. DM is thankful for the JC Bose National Fellowship from SERB, Government of India.

Materials

Acrylamamide Biorad, USA 1610107
Agarose G-Biosciences, USA RC1013
Ammonium persulphate Sigma-Aldrich, USA A3678
anti-ATF4 antibody Cell Signaling Technology, USA 11815 Western blot detection Dilution-1:1000
anti-ATF6 antibody Abcam, UK ab122897 Western blot detection Dilution-1:1000
anti-CHOP antibody Cell Signaling Technology, USA 2897 Western blot detection Dilution-1:1000
anti-eIF2α antibody Santa Cruz Biotechnology, USA sc-11386 Western blot detection Dilution-1:2000 
anti-GADD34 antibody Abcam, UK ab236516 Western blot detection Dilution-1:1000
anti-GAPDH antibody Santa Cruz Biotechnology, USA sc-32233 Western blot detection Dilution-1:3000 
anti-HSPA5 antibody Cell Signaling Technology, USA 3177 Western blot detection Dilution-1:1000
anti-IRE1 antibody Cell Signaling Technology, USA 3294 Western blot detection Dilution-1:2000
Anti-mouse HRP conjugate antibody  Biorad, USA 1706516 Western blot detection Dilution- 1:4000
anti-peIF2α antibody Invitrogen, USA 44-728G Western blot detection Dilution-1:1000
anti-PERK antibody Cell Signaling Technology, USA 5683 Western blot detection Dilution-1:2000
anti-pIRE1 antibody Abcam, UK ab243665 Western blot detection Dilution-1:1000
anti-pPERK antibody Invitrogen, USA PA5-40294 Western blot detection Dilution-1:2000
Anti-rabbit HRP conjugate antibody Biorad, USA 1706515 Western blot detection Dilution- 1:4000
anti-XBP1 antibody Abcam, UK ab37152 Western blot detection Dilution-1:1000
Bench top high speed centrifuge Eppendorf, USA 5804R Rotor- F-45-30-11
Bench top low speed centrifuge Eppendorf, USA 5702R Rotor- A-4-38
Bis-Acrylamide Biorad, USA 1610201
Bovine Serum Albumin (BSA) MP biomedicals, USA 160069
Bradford reagent Biorad, USA 5000006
CalPhos mammalian Transfection kit Clontech, Takara Bio, USA 631312 Virus stock preparation
CEM-GFP NIH, AIDS Repository, USA 3655
Clarity ECL substrate Biorad, USA 1705061 chemiluminescence detecting substrate
Clarity max ECL substrate Biorad, USA 1705062 chemiluminescence detecting substrate
Confocal laser scanning microscope Olympus, Japan Model:FV3000
Cytospin centrifuge Thermo Fisher Scientific, USA ASHA78300003
DMEM Invitrogen, USA 11995073
DMSO Sigma-Aldrich, USA D2650
dNTPs Promega, USA U1515
DTT Invitrogen, USA R0861
EDTA Invitrogen, USA 12635
EtBr Invitrogen, USA `15585011
Fetal Bovine Serum Invitrogen, USA 16000044
G418 Invitrogen, USA 11811023
Glutaraldehyde 25% Sigma-Aldrich, USA G6257 Infectivity assay
Glycine Thermo Fisher Scientific, USA Q24755
HEK-293T NCCS, India
HIV-1 infectious Molecular Clone pNL4-3 NIH, AIDS Repository, USA 114
Inverted microscope Nikon, Japan Model: Eclipse Ti2
iTaq Universal SYBR Green Supermix Biorad, USA 1715124
Jurkat J6 NCCS, India
Magnesium chloride Sigma-Aldrich, USA M8266 Infectivity assay
MMLV-RT  Invitrogen, USA 28025013
MTT reagent Sigma-Aldrich, USA M5655 Cell viability assay
N,N-dimethyl formamide Fluka Chemika 40255 Infectivity assay
NaCl Thermo Fisher Scientific, USA Q27605
NaF Sigma-Aldrich, USA 201154
NP40 Invitrogen, USA 85124
P24 antigen capture ELISA kit ABL, USA 5421
PageRuler prestained protein ladder Sci-fi Biologicals, India PGPMT078
Paraformaldehyde Sigma-Aldrich, USA P6148
pEGFP-N1 Clontech, USA 632515
Penicillin/Streptomycin Invitrogen, USA 151140122
Phosphatase Inhibitor Sigma-Aldrich, USA 4906837001
Phusion High-fidelity PCR mastermix with GC buffer NEB,USA M05532
pLKO.1-TRC Addgene, USA 10878 Lentiviral cloning vector
pMD2.G Addgene, USA 12259 VSV-G envelope vector
PMSF Sigma-Aldrich, USA P7626
Polyethylenimine (PEI)  Polysciences, Inc., USA 23966
Potassium ferricyanide Sigma-Aldrich, USA 244023 Infectivity assay
Potassium ferrocyanide Sigma-Aldrich, USA P3289 Infectivity assay
Protease Inhibitor Sigma-Aldrich, USA  5056489001
psPAX2 Addgene, USA 12260 Lentiviral packaging plasmid
Puromycin Sigma-Aldrich, USA P8833 Selection of stable cells
PVDF membrane Biorad, USA 1620177
Random primers Invitrogen, USA 48190011
RPMI 1640 Invitrogen, USA 22400105
SDS Sigma-Aldrich, USA L3771
Steady-Glo substrate Promega, USA E2510 Luciferase assay
T4 DNA ligase Invitrogen, USA 15224017
TEMED Invitrogen, USA 17919
Thapsigargin Sigma-Aldrich, USA T9033
Thioflavin T Sigma-Aldrich, USA 596200
Tris Thermo Fisher Scientific, USA Q15965
Triton-X-100 Sigma-Aldrich, USA T8787
Trizol Invitrogen, USA 15596018
Tween 20 Sigma-Aldrich, USA P1379
TZM-bl NIH, AIDS Repository, USA 8129
Ultracentrifuge Beckman Optima L90K, USA 330049 Rotor-SW28Ti
UltraPure X-gal Invitrogen, USA 15520-018 Infectivity assay

References

  1. Levy, J. A. . HIV and the Pathogenesis of AIDS. , (2007).
  2. Hebert, D. N., Molinari, M. In and out of the ER: Protein folding, quality control, degradation, and related human diseases. Physiol Rev. 87 (4), 1377-1408 (2007).
  3. Mori, K. The unfolded protein response: The dawn of a new field. Proc Jpn Acad Ser B Phys Biol Sci. 91 (9), 469-480 (2015).
  4. Balch, W. E., Morimoto, R. I., Dillin, A., Kelly, J. W. Adapting proteostasis for disease intervention. Science. 319 (5865), 916-919 (2008).
  5. Kaufman, R. J. Orchestrating the unfolded protein response in health and disease. J Clin Invest. 110 (10), 1389-1398 (2002).
  6. Ron, D., Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 8 (7), 519-529 (2007).
  7. Marciniak, S. J., Garcia-Bonilla, L., Hu, J., Harding, H. P., Ron, D. Activation-dependent substrate recruitment by the eukaryotic translation initiation factor 2 kinase PERK. J Cell Biol. 172 (2), 201-209 (2006).
  8. Harding, H. P., Zhang, Y., Ron, D. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature. 397 (6716), 271-274 (1999).
  9. Vattem, K. M., Wek, R. C. Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells. Proc Natl Acad Sci U S A. 101 (31), 11269-11274 (2004).
  10. Harding, H. P., et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 11 (3), 619-633 (2003).
  11. Novoa, I., Zeng, H., Harding, H. P., Ron, D. Feedback inhibition of the unfolded protein response by GADD34-mediated dephosphorylation of eIF2α. J Cell Biol. 153 (5), 1011-1021 (2001).
  12. Haze, K., Yoshida, H., Yanagi, H., Yura, T., Mori, K. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell. 10 (11), 3787-3799 (1999).
  13. Ye, J., et al. ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell. 6 (6), 1355-1364 (2000).
  14. Tirasophon, W., Welihinda, A. A., Kaufman, R. J. A stress response pathway from the endoplasmic reticulum to the nucleus requires a novel bifunctional protein kinase/endoribonuclease (Ire1p) in mammalian cells. Genes Dev. 12 (12), 1812-1824 (1998).
  15. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T., Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell. 107 (7), 881-891 (2001).
  16. Calfon, M., et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 415 (6867), 92-96 (2002).
  17. Wu, J., et al. ATF6α optimizes long-term endoplasmic reticulum function to protect cells from chronic stress. Dev Cell. 13 (3), 351-364 (2007).
  18. Shoulders, M. D., et al. Stress-independent activation of XBP1s and/or ATF6 reveals three functionally diverse ER proteostasis environments. Cell Rep. 3 (4), 1279-1292 (2013).
  19. Oslowski, C. M., Urano, F. Measuring ER stress and the unfolded protein response using mammalian tissue culture system. Methods Enzymol. 490, 71-92 (2011).
  20. Gupta, S., Samali, A., Fitzgerald, U., Deegan, S. Methods for monitoring endoplasmic reticulum stress and the unfolded protein response. Int J Cell Biol. 2010, 830307 (2010).
  21. Wang, M., Kaufman, R. J. Protein misfolding in the endoplasmic reticulum as a conduit to human disease. Nature. 529 (7586), 326-335 (2016).
  22. Beriault, D. R., Werstuck, G. H. Detection and quantification of endoplasmic reticulum stress in living cells using the fluorescent compound, Thioflavin T. Biochim Biophys Acta. 1833 (10), 2293-2301 (2013).
  23. Tripathi, A., Iyer, K., Mitra, D. HIV-1 replication requires optimal activation of the unfolded protein response. FEBS Lett. 597 (23), 2908-2930 (2023).
  24. Platt, E. J., Wehrly, K., Kuhmann, S. E., Chesebro, B., Kabat, D. Effects of CCR5 and CD4 cell surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus type 1. J Virol. 72 (4), 2855 (1998).
  25. Gervaix, A., West, D., Leoni, L. M., Richman, D. D., Wong-Staal, F., Corbeil, J. A new reporter cell line to monitor HIV infection and drug susceptibility in vitro. Proc Natl Acad Sci U S A. 94 (9), 4653-4658 (1997).
  26. Augustine, T., et al. Cyclin F/FBXO1 interacts with HIV-1 viral infectivity factor (Vif) and restricts progeny virion infectivity by ubiquitination and proteasomal degradation of vif protein through SCFcyclin F E3 ligase machinery. J Biol Chem. 292 (13), 5349-5363 (2017).
  27. Shihan, M. H., Novo, S. G., Le Marchand, S. J., Wang, Y., Duncan, M. K. A simple method for quantitating confocal fluorescent images. Biochem Biophys Rep. 25, 100916 (2021).
  28. Treiman, M., Caspersen, C., Christensen, S. B. A tool coming of age: Thapsigargin as an inhibitor of sarco-endoplasmic reticulum Ca2+-ATPases. Trends Pharmacol Sci. 19 (4), 131-135 (1998).
  29. Moffat, J., et al. A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell. 124 (6), 1283-1298 (2006).
  30. Sodroski, J., Rosen, C., Goh, W. C., Haseltine, W. A Transcriptional activator protein encoded by the x-lor region of the human T-cell leukemia virus. Science. 228 (4706), 1430-1434 (1985).
  31. Rosen, C. A., Sodroski, J. G., Kettman, R., Haseltine, W. A. Activation of enhancer sequences in type II human T-cell leukemia virus and bovine leukemia virus long terminal repeats by virus-associated trans-acting regulatory factors. J Virol. 57 (3), 738-744 (1986).
  32. Dandekar, D. H., Kumar, M., Ladha, J. S., Ganesh, K. N., Mitra, D. A quantitative method for normalization of transfection efficiency using enhanced green fluorescent protein. Anal Biochem. 342 (2), 341-344 (2005).
  33. Mahmood, T., Yang, P. C. Western blot: Technique, theory, and trouble shooting. N Am J Med Sci. 4 (9), 429-434 (2012).
  34. Ghosh, R., Gilda, J. E., Gomes, A. V. The necessity of and strategies for improving confidence in the accuracy of western blots. Expert Rev Proteomics. 11 (5), 549-560 (2014).
  35. Peña, J., Harris, E. Dengue virus modulates the unfolded protein response in a time-dependent manner. J Biol Chem. 286 (16), 14226-14236 (2011).
  36. Soboleski, M. R., Oaks, J., Halford, W. P. Green fluorescent protein is a quantitative reporter of gene expression in individual eukaryotic cells. FASEB J. 19 (3), 440-442 (2005).
  37. Martinez, Z. S., Castro, E., Seong, C. S., Cerón, M. R., Echegoyen, L., Llano, M. Fullerene derivatives strongly inhibit HIV-1 replication by affecting virus maturation without impairing protease activity. Antimicrob Agents Chemother. 60 (10), 5731-5741 (2016).
  38. Askari, S., Javadpour, P., Rashidi, F. S., Dargahi, L., Kashfi, K., Ghasemi, R. Behavioral and molecular effects of Thapsigargin-induced brain ER- stress: Encompassing inflammation, MAPK, and insulin signaling pathway. Life. 12 (9), 1374 (2022).
  39. Jaskulska, A., Janecka, A. E., Gach-Janczak, K. Thapsigargin-from traditional medicine to anticancer drug. Int J Mol Sci. 22 (1), 4 (2021).
  40. Shaban, M. S., et al. Multi-level inhibition of coronavirus replication by chemical ER stress. Nat Commun. 12 (1), 5536 (2021).
  41. Marciniak, S. J., Chambers, J. E., Ron, D. Pharmacological targeting of endoplasmic reticulum stress in disease. Nat Rev Drug Discov. 21 (2), 115-140 (2022).
  42. Sriburi, R., Jackowski, S., Mori, K., Brewer, J. W. XBP1: A link between the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J Cell Biol. 167 (1), 35-41 (2004).
  43. Siwecka, N., Rozpędek-Kamińska, W., Wawrzynkiewicz, A., Pytel, D., Diehl, J. A., Majsterek, I. The structure, activation and signaling of IRE1 and its role in determining cell fate. Biomedicines. 9 (2), 156 (2021).
This article has been published
Video Coming Soon
Keep me updated:

.

Citer Cet Article
Tripathi, A., Dasgupta, A., Mitra, D. Measuring Endoplasmic Reticulum Stress and Unfolded Protein Response in HIV-1 Infected T-Cells and Analyzing its Role in HIV-1 Replication. J. Vis. Exp. (208), e66522, doi:10.3791/66522 (2024).

View Video