Yingji Chen's research while affiliated with Fudan University and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (17)


PPTC7 acts as an essential co-factor of the SCFFBXL4 ubiquitin ligase complex to restrict BNIP3/BNIP3L-dependent mitophagy
  • Preprint

June 2024

·

3 Reads

Xiayun Xu

·

Yingji Chen

·

·

[...]

·

Mitophagy is a selective process that targets damaged, dysfunctional, or superfluous mitochondria for degradation through autophagy. The SCFFBXL4 ubiquitin ligase complex suppresses basal mitophagy by targeting BNIP3 and BNIP3L, two key mitophagy cargo receptors, for ubiquitin-proteasomal degradation. FBXL4 loss-of-function mutations lead to excessive BNIP3/3L-dependent mitophagy, thereby causing a devasting multi-system disorder called mitochondrial DNA depletion syndrome, type 13 (MTDPS13). PPTC7, a mitochondrial matrix phosphatase, is essential for proper mitochondrial function and biogenesis. Here, we show that a proportion of PPTC7 is located on the outer mitochondrial membrane, where it interacts with FBXL4 and BNIP3/3L. PPTC7 decreases BNIP3/3L protein stability in a protein phosphatase activity-independent manner. Using in vitro cell culture and Pptc7 knockout mice models, we demonstrate that PPTC7 deficiency activates high levels of basal mitophagy in a BNIP3/3L-dependent manner. Mechanistically, PPTC7 facilitates SCFFBXL4-mediated ubiquitin-proteasomal degradation of BNIP3/3L. Overall, these findings establish PPTC7 as an essential co-factor of the SCFFBXL4 complex and a suppressor of BNIP3/3L-dependent mitophagy.

Share

Figure 4. Inhibitor of Bruton's tyrosine kinase (IBTK) deficiency reduces eIF4A1-dependent oncoprotein expression and neoplastic phenotypes in cancer cells. (A) Western blot (WB) analysis of the indicated proteins in the whole cell lysates (WCLs) from parental and IBTK-KO SiHa cells (left panel) and the WCLs from SiHa cells treated with silvestrol (100 nM) for the indicated times (right panel). (B) Quantitative reverse transcription polymerase chain reaction (RT-qPCR) assessment of the mRNA expression of eIF4A1 target genes in parental and IBTK-KO SiHa cells. The mRNA levels of Actin Figure 4 continued on next page
mTORC1/S6K1 signaling promotes sustained oncogenic translation through modulating CRL3IBTK-mediated ubiquitination of eIF4A1 in cancer cells
  • Article
  • Full-text available

May 2024

·

14 Reads

eLife

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of the Cullin 3-RING ubiquitin ligase (CRL3) complex, interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 catalyzed by the CRL3 IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and cervical tumor cell growth both in vivo and in vitro. Moreover, we show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3 IBTK complex and the mTORC1/S6K1 signaling pathway, which is frequently dysregulated in cancer, represents a promising target for anti-cancer therapies.

Download

mTORC1/S6K1 signaling promotes sustained oncogenic translation through modulating CRL3IBTK-mediated non-degradative ubiquitination of eIF4A1

May 2024

·

3 Reads

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of Cullin 3-RING ubiquitin ligase (CRL3) complex, interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 catalyzed by CRL3 IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and tumor cell growth both in vivo and in vitro . Moreover, we show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3 IBTK complex and the mTORC1/S6K1 signaling pathway, which is frequently dysregulated in cancer, represents a promising target for anti-cancer therapies. Statement of Significance: Overexpression of IBTK contributes to the process of tumorigenesis by amplifying translation, and represents a promising target for anti-cancer therapies.


Disruption of WSB2-mediated NOXA Degradation Induces Synthetic Lethality to Anti-apoptotic BCL-2 Family Protein Inhibitors

April 2024

·

7 Reads

Anti-apoptotic BCL-2 family proteins are frequently overexpressed in various cancers, contributing to the initiation and development of cancer, as well as intrinsic or acquired resistance to therapy. Although BCL-2 family protein inhibitors, such as Venetoclax, have demonstrated efficacy in hematological neoplasms, their effectiveness as single agents in solid tumors is limited. Identifying alternative molecular targets that can overcome intrinsic resistance to BCL-2 family protein inhibitors is of great clinical importance. Here, we present evidence of strong synthetic lethal interactions between WSB2, a relatively unexplored substrate-binding receptor of the Cullin 5-RBX2-Elongin B/C (CRL5) E3 ubiquitin ligase complex, and multiple anti-apoptotic BCL-2 family proteins. Mechanistically, an assembled CRL5 WSB2 E3 ubiquitin ligase complex targets NOXA, a pro-apoptotic BCL-2 family protein, for degradation via the ubiquitin-proteasomal pathway. Ablation of WSB2 leads to a remarkable accumulation of NOXA proteins in cultured cell lines and knockout mouse organs. While WSB2 deficiency alone has a minimal effect on spontaneous apoptosis, it renders cancer cells more susceptible to apoptosis when anti-apoptotic BCL-2 family proteins are genetically depleted or pharmacologically inhibited. These findings establish WSB2 as a critical regulator of mitochondrial apoptosis and highlight the dysregulation of the WSB2-NOXA regulatory axis as a contributing factor to apoptosis resistance in cancer cells. Synergistically targeting WSB2 and anti-apoptotic BCL-2 family proteins holds promising clinical potential in the treatment of human cancers.


Figure 1. IBTK interacts with eIF4A1 in cells. (A) WB analysis of the indicated proteins in WCLs from FLAG-BirA*-IBTK Tet-on-inducible Flp-In T-REx 293 cells treated with DOX (10 ng/ml) for the indicated times. (B) Affinity purification of IBTK-containing protein complex using FLAG-M2 beads was conducted in FLAG-BirA*-IBTK Tet-oninducible Flp-In T-REx 293 cells. The associated proteins were separated by SDS-PAGE and visualized by Coomassie Blue (CB) staining. (C)The eIF interactome of IBTK identified by AP-MS analysis in (B). The full interaction partner list was showed in Supplementary Table 1. (D-F) WB analysis of the indicated proteins in WCLs and co-IP samples of anti-FLAG antibody obtained from 293T cells transfected with the indicated plasmids. (G) FLAG-BirA*-IBTK Tet-on-inducible Flp-In T-REx 293 cells were treated with/without DOX (10 ng/ml) for 12 h then collected and subjected to co-IP with anti-FLAG antibody. WCLs and co-IP samples were prepared for WB analysis with the indicated antibodies. (H-K) Co-IP using anti-eIF4A1(H), eIF4A2 (I), eIF4A3 (J), or IBTK (K) antibodies in WCLs prepared from 293T cells, followed by WB analysis with the indicated antibodies.
Figure 2. IBTK promotes non-degradative ubiquitination of eIF4A1. (A) WB analysis of the indicated proteins in WCLs from FLAG-BirA*-IBTK Tet-on-inducible T-REx 293 cells treated with DOX (10 ng/ml) for the indicated times. (B) WB analysis of the indicated proteins in WCLs from SiHa and HeLa cells infected with lentivirus expressing IBTK-specific shRNA (#1, #2) or negative control (NC). (C) WB analysis of the indicated proteins in WCLs from parental/IBTK-KO SiHa, 293T, and H1299 cells. (D) WB analysis of the products of in vivo ubiquitination assays from 293T cells transfected with the indicated plasmids. (E) Parental and IBTK-KO 293T cells were transfected with HA-Ub for 24 h, then WCLs were prepared for co-IP with IgG, antieIF4A1, eIF4A2, or eIF4A3 antibodies. The polyubiquitinated forms of eIF4A1/2/3 were detected by WB with anti-HA antibody. (F) WB analysis of the products of in vitro ubiquitination assays with anti-eIF4A1 antibody. (G) WB analysis of the products of in vivo ubiquitination assays from 293T cells transfected with the indicated plasmids.
mTORC1/S6K1 signaling promotes sustained oncogenic translation through modulating CRL3IBTK-mediated non-degradative ubiquitination of eIF4A1

December 2023

·

14 Reads

eLife

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of Culllin 3-RING ubiquitin ligase complex (CRL3), interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 by catalyzed CRL3IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and tumor cell growth both in vivo and in vitro. Moreover, our results show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3IBTK complex and the mTOR signaling pathway, frequently dysregulated in cancer, represents a promising target for anticancer therapies. IBTK overexpression contributes to cervical cancer tumorigenesis by translation regulation and represents a promising target for anticancer therapies.


mTORC1/S6K1 signaling promotes sustained oncogenic translation through modulating CRL3IBTK-mediated non-degradative ubiquitination of eIF4A1

December 2023

·

7 Reads

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of Culllin 3-RING ubiquitin ligase complex (CRL3), interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 by catalyzed CRL3IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and tumor cell growth both in vivo and in vitro. Moreover, our results show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3IBTK complex and the mTOR signaling pathway, frequently dysregulated in cancer, represents a promising target for anticancer therapies. IBTK overexpression contributes to cervical cancer tumorigenesis by translation regulation and represents a promising target for anticancer therapies.


Excessive BNIP3- and BNIP3L-dependent mitophagy underlies the pathogenesis of FBXL4-mutated mitochondrial DNA depletion syndrome

October 2023

·

4 Reads

Mitophagy, the process of removing damaged mitochondria to promote cell survival, plays a crucial role in cellular functionality. However, excessive, or uncontrolled mitophagy can lead to reduced mitochondrial content that burdens the remaining organelles, triggering mitophagy-mediated cell death. FBXL4 mutations, which affect the substrate-binding adaptor of the CUL1 (cullin 1)-RING ubiquitin ligase complex (CRL1), have been linked to mitochondrial DNA depletion syndrome type 13 (MTDPS13) characterized by reduced mtDNA content and impaired energy production in affected organs. However, the mechanism behind FBXL4 mutation-driven MTDPS13 remain poorly understood. In a recent study, we demonstrate that the CRL1-FBXL4 complex promotes the degradation of BNIP3 and BNIP3L, two key mitophagy cargo receptors. Deficiency of FBXL4 results in a strong accumulation of BNIP3 and BNIP3L proteins and triggers high levels of BNIP3- and BNIP3L-dependent mitophagy. Patient-derived FBXL4 mutations do not affect its interaction with BNIP3 and BNIP3L but impair the assembly of an active CRL1-FBXL4 complex. Furthermore, excessive mitophagy is observed in knockin mice carrying a patient-derived FBXL4 mutation, and in cortical neurons generated from human patient induced pluripotent stem cells (hiPSCs). These findings support the model that the CRL1-FBXL4 complex tightly restricts basal mitophagy, and its dysregulation leads to severe symptoms of MTDPS13.


mTORC1/S6K1 signaling promotes sustained oncogenic translation through modulating CRL3IBTK-mediated non-degradative ubiquitination of eIF4A1

September 2023

·

24 Reads

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of Culllin 3-RING ubiquitin ligase complex (CRL3), interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 by catalyzed CRL3IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and tumor cell growth both in vivo and in vitro. Moreover, our results show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3IBTK complex and the mTOR signaling pathway, frequently dysregulated in cancer, represents a promising target for anticancer therapies. Statement of Significance: IBTK overexpression contributes to cervical cancer tumorigenesis by translation regulation and represents a promising target for anticancer therapies.


DPP9 Stabilizes NRF2 to Suppress Ferroptosis and Induce Sorafenib Resistance in Clear Cell Renal Cell Carcinoma

September 2023

·

115 Reads

·

11 Citations

Cancer Research

The KEAP1-NRF2 axis is the principal regulator of cellular responses to oxidative and electrophilic stressors. NRF2 hyperactivation is frequently observed in many types of cancer and promotes cancer initiation, progression, metastasis, and resistance to various therapies. Here, we determined that dipeptidyl peptidase 9 (DPP9) is a regulator of the KEAP1-NRF2 pathway in clear cell renal cell carcinoma (ccRCC). DPP9 was markedly overexpressed at the mRNA and protein levels in ccRCC, and high DPP9 expression levels correlated with advanced tumor stage and poor prognosis in patients with ccRCC. Protein affinity purification to identify functional partners of DPP9 revealed that it bound to KEAP1 via a conserved ESGE motif. DPP9 disrupted KEAP1-NRF2 binding by competing with NRF2 for binding to KEAP1 in an enzyme-independent manner. Upregulation of DPP9 led to stabilization of NRF2, driving NRF2-dependent transcription and thereby decreasing cellular reactive oxygen species levels. Moreover, DPP9 overexpression suppressed ferroptosis and induced resistance to sorafenib in ccRCC cells, which was largely dependent on the NRF2 transcriptional target SLC7A11. Collectively, these findings indicate that the accumulation of DPP9 results in hyperactivation of the NRF2 pathway to promote tumorigenesis and intrinsic drug resistance in ccRCC. Significance DPP9 overcomes oxidative stress and suppresses ferroptosis in ccRCC by binding to KEAP1 and promoting NRF2 stability, which drives tumor development and sorafenib resistance.


Fig. 5. Oxidative/electrophilic stresses potentiate the mTORC2-AKT pathway through inactivating Keap1. (A) WB analysis of the indicated proteins in Ni-NTA pull-downs products and WCL from 293T cells transfected with the indicated plasmids and treated with DMSO,
Fig. 6. EGF-induced intracellular ROS burst mitigates Keap1-mediated mLST8 ubiquitination. (A, B) WB analysis of the indicated proteins in WCL of serum-starved H1299 cells pretreated with DPI (10 μM) for 30 min prior to EGF (100 ng/ml) treatment for the indicated times. At each time point, the relative cellular ROS levels for each condition were measured and shown in (B), the P values are calculated by the Two-way ANOVA test.
Disruption of the Keap1-mTORC2 axis by cancer-derived Keap1/mLST8 mutations leads to oncogenic mTORC2-AKT activation

September 2023

·

26 Reads

·

2 Citations

Redox Biology

The mechanistic target of the rapamycin (mTOR) pathway, which participates in the regulation of cellular growth and metabolism, is aberrantly regulated in various cancer types. The mTOR complex 2 (mTORC2), which consists of the core components mTOR, Rictor, mSin1, and mLST8, primarily responds to growth signals. However, the coordination between mTORC2 assembly and activity remains poorly understood. Keap1, a major sensor of oxidative stress in cells, functions as a substrate adaptor for Cullin 3-RING E3 ubiquitin ligase (CRL3) to promote proteasomal degradation of NF-E2-related factor 2 (NRF2), which is a transcription factor that protects cells against oxidative and electrophilic stress. In the present study, we demonstrate that Keap1 binds to mLST8 via a conserved ETGE motif. The CRL3Keap1 ubiquitin ligase complex promotes non-degradative ubiquitination of mLST8, thus reducing mTORC2 complex integrity and mTORC2-AKT activation. However, this effect can be prevented by oxidative/electrophilic stresses and growth factor signaling-induced reactive oxygen species (ROS) burst. Cancer-derived Keap1 or mLST8 mutations disrupt the Keap1-mLST8 interaction and allow mLST8 to evade Keap1-mediated ubiquitination, thereby enhancing mTORC2-AKT activation and promoting cell malignancy and remodeling cell metabolism. Our findings provide new insights into the molecular mechanisms of Keap1/mLST8 mutation-driven tumorigenesis by promoting mTORC2-AKT activation, which is independent of the canonical NRF2 pathway.


Citations (8)


... In addition, targeting key enzymes in the transsulfuration pathway, such as adenosylhomocysteinase (AHCY; also known as SAHH), cystathionine β-synthase (CBS), and glycine N-methyltransferase (GNMT), can resensitize tumors to ferroptosis inducers targeting SLC7A11 [315][316][317]. In addition, sorafenib, a frontline treatment for advanced hepatocellular carcinoma, has demonstrated ferroptosis induction capabilities in liver, kidney [318] and pancreas by inhibiting system xc − activity [319]. However, the efficacy of sorafenib is limited by acquired drug resistance associated with MT1G expression. ...

Reference:

Ferroptosis: principles and significance in health and disease
DPP9 Stabilizes NRF2 to Suppress Ferroptosis and Induce Sorafenib Resistance in Clear Cell Renal Cell Carcinoma
  • Citing Article
  • September 2023

Cancer Research

... In contrast, genetic alterations in pathways that control CMA, mentioned above, are frequently found in cancer. These include somatic NRF2 and KEAP1 mutations, hypermethylation of KEAP1 and amplification of NRF2, which culminates in a constitutive NRF2 activation; gain-of-function mutations of the canonical transient receptor potential channel (TRPC6) that leads to enhanced NFAT signaling; mutations or overexpression of genes that regulate p38 MAPK activity; chromosomal translocations involving the RARα locus; and mutations on a subunit required for mTORC2 activity (mLST8) that leads to oncogenic mTORC2-AKT activation (Parrado et al., 2000;Pan et al., 2013;Pouremamali et al., 2022;Chen et al., 2023). However, even though mutations in these pathways have the potential to consequently lead to alterations in CMA activity, there is still no clear evidence linking these mutations to their impact on CMA in cancer cells. ...

Disruption of the Keap1-mTORC2 axis by cancer-derived Keap1/mLST8 mutations leads to oncogenic mTORC2-AKT activation

Redox Biology

... Given the post-mitotic nature of neurons, an excess in mitochondrial degradation could be as detrimental as insufficient activation. This is exemplified by disease-causing mutations in FBXL4, which results in excessive mitochondrial degradation through the NIX/BNIP3 pathway [66][67][68][69] and leads to severe mitochondrial encephalopathy 70,71 . Conversely, in cells expressing both mitophagy receptors OPTN and NDP52, NAP1/SIN-TBAD initially compete with OPTN for TBK1 binding until mitophagy is adequately activated. ...

FBXL4 mutations cause excessive mitophagy via BNIP3/BNIP3L accumulation leading to mitochondrial DNA depletion syndrome
  • Citing Article
  • August 2023

Cell Death and Differentiation

... Furthermore, SPOP has been found to act as a tumor suppressor in gastric [29], colorectal [30], and liver cancer [31]. Additionally, it has been found that SPOP mediated the non-degradative ubiquitination of BRAF, leading to inhibition of the MAPK/ERK pathway, and ultimately suppressing tumor development [32]. Our previous study has found SPOP induced non-degradable ubiquitination of Nogo-B and impeded Nogo-B/c-FLIP complex, contributing to suppression of tumor proliferation [31]. ...

SPOP inhibits BRAF-dependent tumorigenesis through promoting non-degradative ubiquitination of BRAF

Cell & Bioscience

... Known substrates of CRL4 DCAF12 Cullin-RING E3 ligase include CCT5, a member of the T-complex protein ring (TRiC) chaperonin complex ( 68 ), as well as the MAGE-A3 and MAGE-A6 melanoma-associated antigens, involved in the regulation of ( 68 ). However , other , yet-unknown recognition modes might also exist for DCAF12, explaining its interaction with IAPs helping cell survival ( 70 ) or MOV10 involved in multiple cellular roles ( 71 ). ...

DCAF12 promotes apoptosis and inhibits NF-κB activation by acting as an endogenous antagonist of IAPs

Oncogene

... KLHDC3 is translocated to the nucleus with c-Myc Whereas c-Myc is localized predominantly in the nucleus, KLHDC3 was previously shown to be present in the cytoplasm [34,35]. We investigated this apparent inconsistency in the subcellular localization of the two proteins by immunocytofluorescence analysis. ...

CRL2-KLHDC3 E3 ubiquitin ligase complex suppresses ferroptosis through promoting p14ARF degradation
  • Citing Article
  • November 2021

Cell Death and Differentiation

... Recently, several reports indicate that STAT5 participates in metabolism regulation [16,48,[57][58][59], and our workgroup has demonstrated that cervical cancer cells express STAT5, and its phosphorylation increased in response to IL-2 [23,24]. Therefore, we used RNAi to knock down STAT5 to analyze its role in metabolism regulation ( Figure 8). ...

Mitochondrial STAT5A promotes metabolic remodeling and the Warburg effect by inactivating the pyruvate dehydrogenase complex

Cell Death and Disease

... SGs could be induced via two pathways that mediate the stress-induced suppression of translation initiation. The first one is the phosphorylation of the α subunit of eukaryotic tribute to drug resistance and compromised therapeutic outcomes [1,[26][27][28][29][30][31][32]. The inhibition of SGs by targeting eIF2α kinases HRI or PERK could improve the sensitivity of cancer cells to SG-inducing anticancer drugs [26,29,32], indicating that SG inhibitors could be employed as a therapeutic strategy to promote the efficacy of cancer treatment and alleviate anticancer drug resistance. ...

Prostate Cancer-associated SPOP mutations enhance cancer cell survival and docetaxel resistance by upregulating Caprin1-dependent stress granule assembly

Molecular Cancer