Energy stress, such as ischemia, induces mitochondrial damage and death in the heart. Degradation of damaged mitochondria by mitophagy is essential for the maintenance of healthy mitochondria and survival. Here we show that mitophagy during myocardial ischemia was mediated predominantly through autophagy characterized by Rab9-associated autophagosomes, rather than the well-characterized form of autophagy that is dependent upon the Atg-conjugation system and LC3. This form of mitophagy played an essential role in protecting the heart against ischemia and was mediated by a protein complex consisting of Ulk1, Rab9, Rip1 and Drp1. This complex allowed recruitment of trans-Golgi membranes associated with Rab9 to damaged mitochondria through Ser179 phosphorylation of Rab9 by Ulk1 and Ser616 phosphorylation of Drp1 by Rip1. Knock-in of Rab9 (S179A) abolished mitophagy and exacerbated injury in response to myocardial ischemia without affecting conventional autophagy. Mitophagy mediated through the Ulk1-Rab9-Rip1-Drp1 pathway protected the heart against ischemia by maintaining healthy mitochondria.
Toshiro Saito, Jihoon Nah, Shin-ichi Oka, Risa Mukai, Yoshiya Monden, Yusuhiro Maejima, Yoshiyuki Ikeda, Sebastiano Sciarretta, Tong Liu, Hong Li, Erdene Baljinnyam, Diego Fraidenraich, Luke Fritzky, Peiyong Zhai, Shizuko Ichinose, Mitsuaki Isobe, Chiao-Po Hsu, Mondira Kundu, Junichi Sadoshima
Peroxisomes perform essential functions in lipid metabolism, including fatty acid oxidation and plasmalogen synthesis. Here, we describe a role for peroxisomal lipid metabolism in mitochondrial dynamics in brown and beige adipocytes. Adipose tissue peroxisomal biogenesis was induced in response to cold exposure through activation of the thermogenic co-regulator PRDM16. Adipose-specific knockout of the peroxisomal biogenesis factor Pex16 (Pex16-AKO) in mice impaired cold tolerance, decreased energy expenditure, and increased diet-induced obesity. Pex16 deficiency blocked cold-induced mitochondrial fission, decreased mitochondrial copy number, and caused mitochondrial dysfunction. Adipose-specific knockout of the peroxisomal beta-oxidation enzyme acyl CoA oxidase 1 (Acox1-AKO) was not sufficient to affect adiposity, thermogenesis or mitochondrial copy number, but knockdown of the plasmalogen synthetic enzyme glyceronephosphate O-acyltransferase (GNPAT) recapitulated the effects of Pex16 inactivation on mitochondrial morphology and function. Plasmalogens are present in mitochondria and decreased with Pex16 inactivation. Their dietary supplementation increased mitochondrial copy number, improved mitochondrial function, and rescued thermogenesis in Pex16-AKO mice. These findings support a surprising interaction between peroxisomes and mitochondria to regulate mitochondrial dynamics and thermogenesis.
Hongsuk Park, Anyuan He, Min Tan, Jordan M. Johnson, John M. Dean, Terri A. Pietka, Yali Chen, Xiangyu Zhang, Fong-Fu Hsu, Babak Razani, Katsuhiko Funai, Irfan J. Lodhi
The adenomatous polyposis coli (APC) gene plays a pivotal role in the pathogenesis of colorectal carcinoma (CRC), but remains a challenge for drug development. Long non-coding RNAs (lncRNAs) are invaluable in identifying cancer pathologies, and providing therapeutic options for cancer patients. Here, we identified a lncRNA (lncRNA-APC1) activated by APC through lncRNA microarray screening, and examined its expression among a large cohort of CRC tissues. A decrease in lncRNA-APC1 expression was positively associated with lymph node and/or distant metastasis, a more advanced clinical stage, as well as a poor prognosis of CRC patients. Additionally, APC can enhance lncRNA-APC1 expression by suppressing the enrichment of PPARα on the lncRNA-APC1 promoter. Furthermore, enforced lncRNA-APC1 expression was sufficient to inhibit CRC cell growth, metastasis and tumor angiogenesis by suppressing exosome production through directly binding Rab5b mRNA and reducing its stability. Importantly, exosomes derived from lncRNA-APC1-silenced CRC cells promoted angiogenesis by activating the MAPK pathway in endothelial cells, and moreover, exosomal Wnt1 largely enhanced CRC cell proliferation and migration through non-canonicial Wnt signaling. Collectively, lncRNA-APC1 is a critical lncRNA regulated by APC in the pathogenesis of CRC. Our findings suggest an APC-regulated lncRNA-APC1 program as an exploitable therapeutic maneuver for CRC patients.
Feng-Wei Wang, Chen-Hui Cao, Kai Han, Yong-Xiang Zhao, Mu-Yan Cai, Zhi-Cheng Xiang, Jia-Xing Zhang, Jie-Wei Chen, Li-Ping Zhong, Yong Huang, Su-Fang Zhou, Xiao-Han Jin, Xin-Yuan Guan, Rui-Hua Xu, Dan Xie
Prostate cancer (PCa) progressed to castration resistance (CRPC) is a fatal disease. CRPC tumors develop resistance to new-generation anti-androgen enzalutamide through lineage plasticity, characterized by epithelial-mesenchymal transition (EMT) and basal-like phenotype. FOXA1 is a transcription factor essential for epithelial lineage differentiation. Here, we demonstrate that FOXA1 loss leads to remarkable up-regulation of transforming growth factor beta 3 (TGFB3), which encodes a ligand of TGF-β pathway. Mechanistically, this is due to genomic occupancy of FOXA1 on an upstream enhancer of TGFB3 gene to directly inhibit its transcription. Functionally, FOXA1 down-regulation induces TGF-β signaling, EMT, and cell motility, which is effectively blocked by TGF-β receptor I inhibitor Galunisertib (LY2157299). Tissue microarray analysis confirmed reduced levels of FOXA1 protein and a concordant increase in TGF-β signaling, indicated by SMAD2 phosphorylation, in CRPC as compared to primary tumors. Importantly, combinatorial LY2157299 treatment sensitized PCa cells to enzalutamide, leading to synergistic effects in inhibiting cell invasion in vitro and xenograft CRPC tumor growth and metastasis in vivo. Therefore, our study establishes FOXA1 as an important regulator of lineage plasticity mediated in part by TGF-β signaling and supports a novel therapeutic strategy to control lineage switching and potentially extend clinical response to antiandrogen therapies.
Bing Song, Su-Hong Park, Jonathan C. Zhao, Ka-wing Fong, Shangze Li, Yongik Lee, Yeqing A. Yang, Subhasree Sridhar, Xiaodong Lu, Sarki A. Abdulkadir, Robert L. Vessella, Colm Morrissey, Timothy M. Kuzel, William J. Catalona, Ximing J. Yang, Jindan Yu
Loss of phosphatase and tensin homolog (PTEN) represents one hallmark of prostate cancer (PCa). However, restoration of PTEN or inhibition of the activated PI3K-AKT pathway has shown limited success, prompting us to identify obligate targets for disease intervention. We hypothesized that PTEN loss might expose cells to unique epigenetic vulnerabilities. Here, we identified a synthetic lethal relationship between PTEN and BRG1, an ATPase subunit of the SWI/SNF chromatin remodeling complex. Higher BRG1 expression in tumors with low PTEN expression was associated with a worse clinical outcome. Genetically engineered mice (GEMs) and organoid assays confirmed that ablation of PTEN sensitized the cells to BRG1 depletion. Mechanistically, PTEN loss stabilized BRG1 protein through the inhibition of the AKT-GSK3β-FBXW7 axis. Increased BRG1 expression in PTEN-deficient PCa cells led to chromatin remodeling into configurations that drive a protumorigenic transcriptome, causing cells to become further addicted to BRG1. Furthermore, we showed in preclinical models that BRG1 antagonist selectively inhibited the progression of PTEN-deficient prostate tumors. Together, our results highlight the synthetic lethal relationship between PTEN and BRG1, and support targeting BRG1 as an effective approach to the treatment of PTEN-deficient PCa.
Yufeng Ding, Ni Li, Baijun Dong, Wangxin Guo, Hui Wei, Qilong Chen, Huairui Yuan, Ying Han, Hanwen Chang, Shan Kan, Xuege Wang, Qiang Pan, Ping Wu, Chao Peng, Tong Qiu, Qintong Li, Dong Gao, Wei Xue, Jun Qin
Tumor cure with conventional fractionated radiotherapy is 65%, dependent on tumor cell-autonomous gradual buildup of DNA double strand break (DSB) misrepair. Here we report single dose radiotherapy (SDRT), a disruptive technique that ablates >90% of human cancers, operates a distinct dual-target mechanism, linking acid sphingomyelinase (ASMase)-mediated microvascular perfusion defects to DNA unrepair in tumor cells to confer tumor cell lethality. ASMase-mediated microcirculatory vasoconstriction post-SDRT conferred an ischemic stress response within parenchymal tumor cells, with reactive oxygen species triggering the evolutionarily conserved SUMO Stress Response, specifically depleting chromatin-associated free SUMO3. Whereas SUMO3, but not SUMO2, was indispensible for homology-directed repair (HDR) of DSBs, HDR loss-of-function post-SDRT yielded DSB unrepair, chromosomal aberrations and tumor clonogen demise. Vasoconstriction blockade with the endothelin-1 inhibitor BQ-123, or ROS scavenging post-SDRT using peroxiredoxin-6 overexpression or the SOD-mimetic tempol, prevented chromatin SUMO3 depletion, HDR loss-of-function and SDRT tumor ablation. We also provide evidence of mouse to human translation of this biology in a randomized clinical trial, showing 24Gy SDRT, but not 3x9Gy fractionation, coupled early tumor ischemia/reperfusion to human cancer ablation. The SDRT biology provides opportunities for mechanism-based selective tumor radiosensitization via accessing SDRT/ASMase signaling, as current studies indicate this pathway is tractable to pharmacologic intervention.
Sahra Bodo, Cecile Campagne, Tin Htwe Thin, Daniel S. Higginson, H. Alberto Vargas, Guoqiang Hua, John D. Fuller, Ellen Ackerstaff, James Russell, Zhigang Zhang, Stefan Klingler, HyungJoon Cho, Matthew G. Kaag, Yousef Mazaheri, Andreas Rimner, Katia Manova-Todorova, Boris Epel, Joan Zatcky, Cristian R. Cleary, Shyam S. Rao, Yoshiya Yamada, Michael J. Zelefsky, Howard J. Halpern, Jason A. Koutcher, Carlos Cordon-Cardo, Carlo Greco, Adriana Haimovitz-Friedman, Evis Sala, Simon N. Powell, Richard Kolesnick, Zvi Fuks
Abnormal alternative splicing (AS) caused by alterations of splicing factors contributes to tumor progression. Serine/arginine splicing factor 1 (SRSF1) has emerged as a key oncodriver in numerous solid tumors, leaving its roles and mechanisms largely obscure in glioma. Herein we demonstrated that SRSF1 was increased in glioma tissues and cell lines. Moreover, its expression was correlated positively with tumor grade and Ki-67 index, but inversely with patients’ survival. Using RNA-seq, we comprehensively screened and identified multiple SRSF1-affected AS events. Motif analysis revealed a position-dependent modulation of AS by SRSF1 in glioma. Functionally, we verified that SRSF1 promoted cell proliferation, survival and invasion by specifically switching the AS of myosin IB (MYO1B) gene and facilitating the expression of the oncogenic and membrane-localized isoform, MYO1B-fl. Strikingly, MYO1B splicing was dysregulated in parallel with SRSF1 expression in gliomas, and predicted the poor prognosis of the patients. Further investigation revealed that SRSF1-guided AS of MYO1B gene increased the tumorigenic potentials of glioma cells through the PDK1/AKT and PAK/LIMK pathways. Taken together, we identify SRSF1 as an important oncodriver, which integrates the AS controlling of MYO1B into promotion of gliomagenesis, and represents a potential prognostic biomarker and target for glioma therapy.
Xuexia Zhou, Run Wang, Xuebing Li, Lin Yu, Dan Hua, Cuiyun Sun, Cuijuan Shi, Wenjun Luo, Chun Rao, Zhendong Jiang, Ying Feng, Qian Wang, Shizhu Yu
Immune checkpoint therapies have shown tremendous promise in cancer therapy. However, tools to assess their target engagement, and hence ability to predict their efficacy, have been lacking. Here, we show that target engagement and tumor residence kinetics of antibody therapeutics targeting the programmed death ligand-1 (PD-L1) can be quantified non-invasively. In computational docking studies, we observed that PD-L1-targeted antibodies (atezolizumab, avelumab, durvalumab) and a high affinity PD-L1 binding peptide, WL12, have common interaction sites on PD-L1. Using the peptide radiotracer [64Cu]WL12 in vivo, we employed positron emission tomography (PET) imaging and biodistribution studies, in multiple xenograft models and demonstrated that variable PD-L1 expression and its saturation by atezolizumab, avelumab, and durvalumab can be quantified independent of biophysical properties and pharmacokinetics of antibodies. Next, we used [64Cu]WL12 to evaluate the impact of time and dose on free fraction of tumor PD-L1 levels during treatment. These quantitative measures enabled, by mathematical modeling, prediction of antibody doses needed to achieve therapeutically effective occupancy (defined as >90%). Thus, we show that peptide-based PET is a promising tool for optimizing dose and therapeutic regimens employing PD-L1 checkpoint antibodies, and can be used for improving therapeutic efficacy.
Dhiraj Kumar, Ala Lisok, Elyes Dahmane, Matthew D. McCoy, Sagar Shelake, Samit Chatterjee, Viola Allaj, Polina Sysa-Shah, Bryan Wharram, Wojciech G. Lesniak, Ellen Tully, Edward Gabrielson, Elizabeth M. Jaffee, John T. Poirier, Charles M. Rudin, Jogarao V.S. Gobburu, Martin G. Pomper, Sridhar Nimmagadda
Macrophages perform key functions in tissue homeostasis that are influenced by the local tissue environment. Within the tumor microenvironment tumor associated macrophages can be altered to acquire properties that enhance tumor growth. Here, we found lactate, a metabolite found in high concentration within the anaerobic tumor environment, activated mTORC1 that subsequently suppressed TFEB-mediated expression of a macrophage-specific vacuolar ATPase subunit ATP6V0d2. Atp6v0d2-/- mice were more susceptible to tumor growth with enhanced HIF-2α-mediated VEGF production in macrophages that display a more protumoral phenotype. We found that ATP6V0d2 targeted HIF-2α but not HIF-1α for lysosome-mediated degradation. Blockade of HIF-2α transcriptional activity reversed the susceptibility of Atp6v0d2-/- mice to tumor development. Furthermore, in a cohort of patients with lung adenocarcinoma, expression of ATP6V0d2 and HIF-2α was positively and negatively correlated with survival respectively, suggesting a critical role of the macrophage lactate-ATP6V0d2-HIF-2α axis in maintaining tumor growth in human patients. Together, our results highlight the ability of tumor cells to modify the function of tumor-infiltrating macrophages to optimize the microenvironment for tumor growth.
Na Liu, Jing Luo, Dong Kuang, Sanpeng Xu, Yaqi Duan, Yu Xia, Zhengping Wei, Xiuxiu Xie, Bingjiao Yin, Fang Chen, Shunqun Luo, Huicheng Liu, Jing Wang, Kan Jiang, Feili Gong, Zhao-hui Tang, Xiang Cheng, Huabin Li, Zhuoya Li, Arian Laurence, Guoping Wang, Xiang-Ping Yang
Ca2+ channel β-subunit interactions with pore-forming α-subunits are long-thought to be obligatory for channel trafficking to the cell surface and for tuning of basal biophysical properties in many tissues. Unexpectedly, we demonstrate that transgenic expression of mutant cardiac α1C subunits lacking capacity to bind CaVβ because of alanine-substitutions of three conserved residues — Y467, W470, and I471 in the α-interaction domain of rabbit α1C — can traffic to the sarcolemma in adult cardiomyocytes in vivo and sustain normal excitation-contraction coupling. However, these β-less Ca2+ channels cannot be stimulated by β-adrenergic pathway agonists, and thus adrenergic-augmentation of contractility is markedly impaired in isolated cardiomyocytes and in hearts. Similarly, viral-mediated expression of a β-subunit-sequestering-peptide sharply curtailed β-adrenergic stimulation of wild-type Ca2+ channels, identifying an approach to specifically modulate β-adrenergic regulation of cardiac contractility. Our data demonstrate that β subunits are required for β-adrenergic regulation of CaV1.2 channels and positive inotropy in the heart, but are dispensable for CaV1.2 trafficking to the adult cardiomyocyte cell surface, and for basal function and excitation-contraction coupling.
Lin Yang, Alexander Katchman, Jared S. Kushner, Alexander Kushnir, Sergey I. Zakharov, Bi-xing Chen, Zunaira Shuja, Prakash Subramanyam, Guoxia Liu, Arianne Papa, Daniel D. Roybal, Geoffrey S. Pitt, Henry M. Colecraft, Steven O. Marx
No posts were found with this tag.