bh3 domain
Recently Published Documents


TOTAL DOCUMENTS

178
(FIVE YEARS 19)

H-INDEX

50
(FIVE YEARS 2)

2021 ◽  
Vol 141 ◽  
pp. 111825
Author(s):  
Zhenqingyun Shuai ◽  
Yongxiang Zheng ◽  
Jia Jiang ◽  
Rong Yu ◽  
Chun Zhang

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Shanming Hu ◽  
Yanan Wang ◽  
Zhengmao Xu ◽  
Yongzhi Zhou ◽  
Jie Cao ◽  
...  

Abstract Background The salivary glands of female ticks degenerate rapidly by apoptosis and autophagy after feeding. Bcl-2 family proteins play an important role in the apoptosis pathways, but the functions of these proteins in ticks are unclear. We studied Bcl-2 and Bax homologs from Rhipicephalus haemaphysaloides and determined their functions in the degeneration of the salivary glands. Methods Two molecules containing conserved BH (Bcl-2 family homology) domains were identified and named RhBcl-2 and RhBax. After protein purification and mouse immunization, specific polyclonal antibodies (PcAb) were created in response to the recombinant proteins. Reverse transcription quantitative PCR (RT-qPCR) and western blot were used to detect the presence of RhBcl-2 and RhBax in ticks. TUNEL assays were used to determine the level of apoptosis in the salivary glands of female ticks at different feeding times after gene silencing. Co-transfection and GST pull-down assays were used to identify interactions between RhBcl-2 and RhBax. Results The RT-qPCR assay revealed that RhBax gene transcription increased significantly during feeding at all tick developmental stages (engorged larvae, nymphs, and adult females). Transcriptional levels of RhBcl-2 and RhBax increased more significantly in the female salivary glands than in other tissues post engorgement. RhBcl-2 silencing significantly inhibited tick feeding. In contrast, RhBax interference had no effect on tick feeding. TUNEL staining showed that apoptosis levels were significantly reduced after interference with RhBcl-2 expression. Co-transfection and GST pull-down assays showed that RhBcl-2 and RhBax could interact but not combine in the absence of the BH3 domain. Conclusions This study identified the roles of RhBcl-2 and RhBax in tick salivary gland degeneration and finds that the BH3 domain is a key factor in their interactions. Graphical Abstract


Oncogenesis ◽  
2021 ◽  
Vol 10 (7) ◽  
Author(s):  
Yongyang Luo ◽  
Hanyong Jin ◽  
Je Hyeong Kim ◽  
Jeehyeon Bae

AbstractInterferon-inducible guanylate-binding proteins (GBPs) are well-known for mediating host-defense mechanisms against cellular pathogens. Emerging evidence suggests that GBPs are also implicated in tumorigenesis; however, their underlying molecular mechanism is still unknown. In this study, we identified that GBP1 and GBP2 interact with MCL-1, the key prosurvival member of the BCL-2 family, via its BH3 domain. GBPs induce caspase-dependent apoptosis in chronic myeloid leukemia (CML) and acute myeloid leukemia (AML) cells, where the proapoptotic BCL-2 member, BAK, is an indispensable mediator. In particular, GBP2 completely inhibited the MCL-1-mediated promotion of the survival of CML cells through competitive inhibition, resulting in BAK liberation from MCL-1. Concurrently, GBP2 dramatically upregulates BAK expression via its inhibition of the PI3K/AKT pathway. Moreover, paclitaxel upregulates GBP2 expression, and paclitaxel-induced apoptotic activity was distinctively compromised by knockout of GBP2 in CML cells. Bioinformatics analyses of leukemia databases revealed that transcripts of GBPs were generally downregulated in leukemia patients and that GBPs were favorable prognosis markers. Thus, these findings provide molecular evidence of GBPs as apoptosis-inducing proteins of leukemia cells and suggest that GBPs are attractive targets for the development of chemotherapeutics.


2021 ◽  
Author(s):  
Logan P. Poole ◽  
Althea Bock-Hughes ◽  
Damian E. Berardi ◽  
Kay F. Macleod

Abstract UNC51-like kinase-1 (ULK1) is the catalytic component of the autophagy pre-initiation complex that stimulates autophagy via phosphorylation of ATG14, BECLN1 and other autophagy proteins. ULK1 has also been shown to specifically promote mitophagy but the mechanistic basis of how has remained unclear. Here we show that ULK1 phosphorylates the BNIP3 mitochondrial cargo receptor on a critical serine residue (S17) adjacent to its amino terminal LIR motif. ULK1 similarly phosphorylates BNIP3L on S35. Phosphorylation of BNIP3 on S17 by ULK1 promotes interaction with LC3 and mitophagy. ULK1 interaction also promotes BNIP3 protein stability by limiting its turnover at the proteasome. The ability of ULK1 to regulate BNIP3 protein stability depends on an intact “BH3” domain and deletion of its “BH3” domain reduces BNIP3 turnover and increases BNIP3 protein levels independent of ULK1. In summary ULK1 promotes mitophagy by both stabilization of BNIP3 protein and via phosphorylation of S17 to stimulate interaction with LC3.


Author(s):  
Chuanliang Zhang ◽  
Lijuan Wu ◽  
Xiaochun Liu ◽  
Jiangming Gao ◽  
Shan Liu ◽  
...  
Keyword(s):  

Pharmaceutics ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 583
Author(s):  
Suzanne I. Sandin ◽  
David M. Gravano ◽  
Christopher J. Randolph ◽  
Meenakshi Sharma ◽  
Eva de Alba

Saposin C (sapC) is a lysosomal, peripheral-membrane protein displaying liposome fusogenic capabilities. Proteoliposomes of sapC and phosphatidylserine have been shown to be toxic for cancer cells and are currently on clinical trial to treat glioblastoma. As proof-of-concept, we show two strategies to enhance the applications of sapC proteoliposomes: (1) Engineering chimeras composed of sapC to modulate proteoliposome function; (2) Engineering sapC to modify its lipid binding capabilities. In the chimera design, sapC is linked to a cell death-inducing peptide: the BH3 domain of the Bcl-2 protein PUMA. We show by solution NMR and dynamic light scattering that the chimera is functional at the molecular level by fusing liposomes and by interacting with prosurvival Bcl-xL, which is PUMA’s known mechanism to induce cell death. Furthermore, sapC-PUMA proteoliposomes enhance cytotoxicity in glioblastoma cells compared to sapC. Finally, the sapC domain of the chimera has been engineered to optimize liposome binding at pH close to physiological values as protein–lipid interactions are favored at acidic pH in the native protein. Altogether, our results indicate that the properties of sapC proteoliposomes can be modified by engineering the protein surface and by the addition of small peptides as fusion constructs.


2021 ◽  
Author(s):  
Shanming Hu ◽  
Yanan Wang ◽  
Zhengmao Xu ◽  
Yongzhi Zhou ◽  
Jie Cao ◽  
...  

Abstract BackgroundThe salivary gland of female ticks degenerates rapidly by apoptosis and autophagy after feeding. Bcl-2 family proteins play an important role in the apoptosis pathways but the functions of these proteins in ticks are unclear. We studied Bcl-2 and Bax homologs from the tick Rhipicephalus haemaphysaloides and determined their functions in the degeneration of the salivary glands.MethodsThe full-length cDNA of the RhBcl-2 and RhBax genes were obtained by transcriptome analysis and RhBcl-2 and RhBax were expressed in E. coli. Two molecules containing conserved BH (Bcl-2 family homology) domains were identified and named RhBcl-2 and RhBax. After protein purification and mouse immunizations, specific polyclonal antibodies (PcAb) were created in response to the recombinant protein. Reverse transcription quantitative PCR (RT-qPCR) and western blot were used to detect the existence of RhBcl-2 and RhBax in ticks. TUNEL assays were used to determine the apoptosis level in the salivary glands at different feeding times after gene silencing. The interactions between RhBcl-2 and RhBax were identified by co-transfection and GST pull-down assays. The RT-qPCR assay demonstrated that the transcription of RhBax genes increased significantly during the engorged periods of the tick developmental stages (engorged larval, nymph, and adult female ticks). ResultsTranscriptional levels of RhBcl-2 and RhBax in the salivary glands increased more significantly than other tissues post engorgement. RhBcl-2 treatment significantly restrained tick feeding. In contrast, RhBax interference had no effect on tick feeding. TUNEL staining showed that apoptosis levels were significantly reduced after interfering with RhBcl-2 expression. Co-transfection and GST pull-down assays showed that RhBcl-2 and RhBax could combine with each other, but failed to combine without the BH3 domain. ConclusionsThis study identified the roles of RhBcl-2 and RhBax in tick salivary gland degeneration and clarifies their interactions.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Yuko Nishiwaki ◽  
Ichiro Masai

Abstract BNip1, which functions as a t-SNARE component of the syntaxin18 complex, is localized on the ER membrane and regulates retrograde transport from Golgi to the ER. BNip1 also has a BH3 domain, which generally releases pro-apoptotic proteins from Bcl2-mediated inhibition. Previously we reported that retinal photoreceptors undergo BNip1-dependent apoptosis in zebrafish β-snap1 mutants. Here, we investigated physiological roles of BNip1-dependent photoreceptor apoptosis. First, we examined the spatio-temporal profile of photoreceptor apoptosis in β-snap1 mutants, and found that apoptosis occurs only during a small developmental window, 2–4 days-post-fertilization (dpf), in which an apical photoreceptive membrane structure, called the outer segment (OS), grows rapidly. Transient expression of β-SNAP1 during this OS growing period prevents photoreceptor apoptosis in β-snap1 mutants, enabling cone to survive until at least 21 dpf. These observations suggest that BNip1-mediated apoptosis is linked to excessive activation of vesicular transport associated with rapid growth of the OS. Consistently, knockdown of Ift88 and Kif3b, which inhibits protein transport to the OS, rescued photoreceptor apoptosis in β-snap1 mutants. Treatment with rapamycin, which inhibits protein synthesis via the mTOR pathway, also rescued photoreceptor apoptosis in β-snap1 mutants. These data suggest that BNip1 performs risk assessment to detect excessive vesicular transport in photoreceptors.


2020 ◽  
Vol 6 (1) ◽  
Author(s):  
Liora Lindenboim ◽  
Dan Grozki ◽  
Ayelet R. Amsalem-Zafran ◽  
Aida Peña-Blanco ◽  
Gregg G. Gundersen ◽  
...  

Abstract The canonical function of Bcl-2 family proteins is to regulate mitochondrial membrane integrity. In response to apoptotic signals the multi-domain pro-apoptotic proteins Bax and Bak are activated and perforate the mitochondrial outer membrane by a mechanism which is inhibited by their interaction with pro-survival members of the family. However, other studies have shown that Bax and Bak may have additional, non-canonical functions, which include stress-induced nuclear envelope rupture and discharge of nuclear proteins into the cytosol. We show here that the apoptotic stimuli cisplatin and staurosporine induce a Bax/Bak-dependent degradation and subcellular redistribution of nesprin-1 and nesprin-2 but not nesprin-3, of the linker of nucleoskeleton and cytoskeleton (LINC) complex. The degradation and redistribution were caspase-independent and did not occur in Bax/Bak double knockout (DKO) mouse embryo fibroblasts (MEFs). Re-expression of Bax in Bax/Bak DKO MEFs restored stress-induced redistribution of nesprin-2 by a mechanism which requires Bax membrane localization and integrity of the α helices 5/6, and the Bcl-2 homology 3 (BH3) domain. We found that nesprin-2 interacts with Bax in close proximity to perinuclear mitochondria in mouse and human cells. This interaction requires the mitochondrial targeting and N-terminal region but not the BH3 domain of Bax. Our results identify nesprin-2 as a Bax binding partner and also a new function of Bax in impairing the integrity of the LINC complex.


Biomolecules ◽  
2020 ◽  
Vol 10 (9) ◽  
pp. 1308
Author(s):  
Arvind Ramanathan ◽  
Akash Parvatikar ◽  
Srinivas C. Chennubhotla ◽  
Yang Mei ◽  
Sangita C. Sinha

Viral BCL2 proteins (vBCL2s) help to sustain chronic infection of host proteins to inhibit apoptosis and autophagy. However, details of conformational changes in vBCL2s that enable binding to BH3Ds remain unknown. Using all-atom, multiple microsecond-long molecular dynamic simulations (totaling 17 μs) of the murine γ-herpesvirus 68 vBCL2 (M11), and statistical inference techniques, we show that regions of M11 transiently unfold and refold upon binding of the BH3D. Further, we show that this partial unfolding/refolding within M11 is mediated by a network of hydrophobic interactions, which includes residues that are 10 Å away from the BH3D binding cleft. We experimentally validate the role of these hydrophobic interactions by quantifying the impact of mutating these residues on binding to the Beclin1/BECN1 BH3D, demonstrating that these mutations adversely affect both protein stability and binding. To our knowledge, this is the first study detailing the binding-associated conformational changes and presence of long-range interactions within vBCL2s.


Sign in / Sign up

Export Citation Format

Share Document