Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Diego Acosta-Alvear is active.

Publication


Featured researches published by Diego Acosta-Alvear.


eLife | 2013

Pharmacological brake-release of mRNA translation enhances cognitive memory.

Carmela Sidrauski; Diego Acosta-Alvear; Arkady Khoutorsky; Punitha Vedantham; Brian R. Hearn; Han Hua Li; Karine Gamache; Ciara M. Gallagher; Kenny K-H Ang; Chris Wilson; Voytek Okreglak; Avi Ashkenazi; Byron Hann; Karim Nader; Michelle R. Arkin; Adam R. Renslo; Nahum Sonenberg; Peter Walter

Phosphorylation of the α-subunit of initiation factor 2 (eIF2) controls protein synthesis by a conserved mechanism. In metazoa, distinct stress conditions activate different eIF2α kinases (PERK, PKR, GCN2, and HRI) that converge on phosphorylating a unique serine in eIF2α. This collection of signaling pathways is termed the ‘integrated stress response’ (ISR). eIF2α phosphorylation diminishes protein synthesis, while allowing preferential translation of some mRNAs. Starting with a cell-based screen for inhibitors of PERK signaling, we identified a small molecule, named ISRIB, that potently (IC50 = 5 nM) reverses the effects of eIF2α phosphorylation. ISRIB reduces the viability of cells subjected to PERK-activation by chronic endoplasmic reticulum stress. eIF2α phosphorylation is implicated in memory consolidation. Remarkably, ISRIB-treated mice display significant enhancement in spatial and fear-associated learning. Thus, memory consolidation is inherently limited by the ISR, and ISRIB releases this brake. As such, ISRIB promises to contribute to our understanding and treatment of cognitive disorders. DOI: http://dx.doi.org/10.7554/eLife.00498.001


Journal of Cell Biology | 2007

Retinoblastoma tumor suppressor protein–dependent methylation of histone H3 lysine 27 is associated with irreversible cell cycle exit

Alexandre Blais; Chris van Oevelen; Raphael Margueron; Diego Acosta-Alvear; Brian David Dynlacht

The retinoblastoma tumor suppressor protein (pRb) is involved in mitotic exit, promoting the arrest of myoblasts, and myogenic differentiation. However, it is unclear how permanent cell cycle exit is maintained in differentiated muscle. Using RNA interference, expression profiling, and chromatin immunoprecipitations, we show that pRb is essential for cell cycle exit and the differentiation of myoblasts and is also uniquely required to maintain this arrest in myotubes. Remarkably, we also uncover a function for the pRb-related proteins p107 and p130 as enforcers of a G2/M phase checkpoint that prevents progression into mitosis in cells that have lost pRb. We further demonstrate that pRb effects permanent cell cycle exit in part by maintaining trimethylation of histone H3 lysine 27 (H3K27) on cell cycle genes. H3K27 trimethylation silences other genes, including Cyclin D1, in a pRb-independent but polycomb-dependent manner. Thus, our data distinguish two distinct chromatin-based regulatory mechanisms that lead to terminal differentiation.


Journal of Cell Biology | 2011

A CHOP-regulated microRNA controls rhodopsin expression

Shannon Behrman; Diego Acosta-Alvear; Peter Walter

ER stress induces expression of miR-708, which suppresses the production of rhodopsin to prevent ER overloading in retinal epithelial cells.


Genes & Development | 2009

E2f3b plays an essential role in myogenic differentiation through isoform-specific gene regulation

Patrik Asp; Diego Acosta-Alvear; Mary Tsikitis; Chris van Oevelen; Brian David Dynlacht

Current models posit that E2F transcription factors can be divided into members that either activate or repress transcription, in part through collaboration with the retinoblastoma (pRb) tumor suppressor family. The E2f3 locus encodes E2f3a and E2f3b proteins, and available data suggest that they regulate cell cycle-dependent gene expression through opposing transcriptional activating and repressing activities in growing and quiescent cells, respectively. However, the role, if any, of E2F proteins, and in particular E2f3, in myogenic differentiation is not well understood. Here, we dissect the contributions of E2f3 isoforms and other activating and repressing E2Fs to cell cycle exit and differentiation by performing genome-wide identification of isoform-specific targets. We show that E2f3a and E2f3b target genes are involved in cell growth, lipid metabolism, and differentiation in an isoform-specific manner. Remarkably, using gene silencing, we show that E2f3b, but not E2f3a or other E2F family members, is required for myogenic differentiation, and that this requirement for E2f3b does not depend on pRb. Our functional studies indicate that E2f3b specifically attenuates expression of genes required to promote differentiation. These data suggest how diverse E2F isoforms encoded by a single locus can play opposing roles in cell cycle exit and differentiation.


Molecular Cancer Therapeutics | 2015

Validation of the Hsp70–Bag3 Protein–Protein Interaction as a Potential Therapeutic Target in Cancer

Xiaokai Li; Teresa Colvin; Jennifer N. Rauch; Diego Acosta-Alvear; Martin Kampmann; Bryan M. Dunyak; Byron Hann; Blake T. Aftab; Megan Murnane; Min Cho; Peter Walter; Jonathan S. Weissman; Michael Y. Sherman; Jason E. Gestwicki

Hsp70 is a stress-inducible molecular chaperone that is required for cancer development at several steps. Targeting the active site of Hsp70 has proven relatively challenging, driving interest in alternative approaches. Hsp70 collaborates with the Bcl2-associated athanogene 3 (Bag3) to promote cell survival through multiple pathways, including FoxM1. Therefore, inhibitors of the Hsp70–Bag3 protein–protein interaction (PPI) may provide a noncanonical way to target this chaperone. We report that JG-98, an allosteric inhibitor of this PPI, indeed has antiproliferative activity (EC50 values between 0.3 and 4 μmol/L) across cancer cell lines from multiple origins. JG-98 destabilized FoxM1 and relieved suppression of downstream effectors, including p21 and p27. On the basis of these findings, JG-98 was evaluated in mice for pharmacokinetics, tolerability, and activity in two xenograft models. The results suggested that the Hsp70–Bag3 interaction may be a promising, new target for anticancer therapy. Mol Cancer Ther; 14(3); 642–8. ©2015 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Targeting IRE1 with small molecules counteracts progression of atherosclerosis

Özlem Tufanlı; Pelin Telkoparan Akillilar; Diego Acosta-Alvear; Begüm Kocatürk; Umut Inci Onat; Syed Muhammad Hamid; Ismail Çimen; Peter Walter; Christian Weber; Ebru Erbay

Significance Endoplasmic reticulum (ER) stress is linked to the development of complex metabolic diseases, including diabetes, obesity, and atherosclerosis. Irremediable ER stress can push the unfolded protein response (UPR) toward proinflammatory and proapoptotic signaling. The need to dissociate the adaptive UPR responses from its destructive outputs has become a major challenge for therapeutic strategies aimed at mitigating ER stress that is often observed in chronic diseases. Our findings show that inositol-requiring enzyme 1 (IRE1) plays a critical role in metaflammation and that administering IRE1-specific inhibitors to hyperlipidemic mice counteracts atherosclerosis progression. Metaflammation, an atypical, metabolically induced, chronic low-grade inflammation, plays an important role in the development of obesity, diabetes, and atherosclerosis. An important primer for metaflammation is the persistent metabolic overloading of the endoplasmic reticulum (ER), leading to its functional impairment. Activation of the unfolded protein response (UPR), a homeostatic regulatory network that responds to ER stress, is a hallmark of all stages of atherosclerotic plaque formation. The most conserved ER-resident UPR regulator, the kinase/endoribonuclease inositol-requiring enzyme 1 (IRE1), is activated in lipid-laden macrophages that infiltrate the atherosclerotic lesions. Using RNA sequencing in macrophages, we discovered that IRE1 regulates the expression of many proatherogenic genes, including several important cytokines and chemokines. We show that IRE1 inhibitors uncouple lipid-induced ER stress from inflammasome activation in both mouse and human macrophages. In vivo, these IRE1 inhibitors led to a significant decrease in hyperlipidemia-induced IL-1β and IL-18 production, lowered T-helper type-1 immune responses, and reduced atherosclerotic plaque size without altering the plasma lipid profiles in apolipoprotein E-deficient mice. These results show that pharmacologic modulation of IRE1 counteracts metaflammation and alleviates atherosclerosis.


eLife | 2017

An unfolded protein-induced conformational switch activates mammalian IRE1

G. Elif Karagöz; Diego Acosta-Alvear; Hieu T. Nguyen; Crystal P. Lee; Feixia Chu; Peter Walter

The unfolded protein response (UPR) adjusts the cell’s protein folding capacity in the endoplasmic reticulum (ER) according to need. IRE1 is the most conserved UPR sensor in eukaryotic cells. It has remained controversial, however, whether mammalian and yeast IRE1 use a common mechanism for ER stress sensing. Here, we show that similar to yeast, human IRE1α’s ER-lumenal domain (hIRE1α LD) binds peptides with a characteristic amino acid bias. Peptides and unfolded proteins bind to hIRE1α LD’s MHC-like groove and induce allosteric changes that lead to its oligomerization. Mutation of a hydrophobic patch at the oligomerization interface decoupled peptide binding to hIRE1α LD from its oligomerization, yet retained peptide-induced allosteric coupling within the domain. Importantly, impairing oligomerization of hIRE1α LD abolished IRE1’s activity in living cells. Our results provide evidence for a unifying mechanism of IRE1 activation that relies on unfolded protein binding-induced oligomerization.


eLife | 2015

Endoplasmic reticulum stress-independent activation of unfolded protein response kinases by a small molecule ATP-mimic

Aaron S Mendez; Jennifer Alfaro; Marisol A Morales-Soto; Arvin C. Dar; Emma McCullagh; Katja Gotthardt; Han Li; Diego Acosta-Alvear; Carmela Sidrauski; Alexei Korennykh; Sebastián Bernales; Kevan M. Shokat; Peter Walter

Two ER membrane-resident transmembrane kinases, IRE1 and PERK, function as stress sensors in the unfolded protein response. IRE1 also has an endoribonuclease activity, which initiates a non-conventional mRNA splicing reaction, while PERK phosphorylates eIF2α. We engineered a potent small molecule, IPA, that binds to IRE1s ATP-binding pocket and predisposes the kinase domain to oligomerization, activating its RNase. IPA also inhibits PERK but, paradoxically, activates it at low concentrations, resulting in a bell-shaped activation profile. We reconstituted IPA-activation of PERK-mediated eIF2α phosphorylation from purified components. We estimate that under conditions of maximal activation less than 15% of PERK molecules in the reaction are occupied by IPA. We propose that IPA binding biases the PERK kinase towards its active conformation, which trans-activates apo-PERK molecules. The mechanism by which partial occupancy with an inhibitor can activate kinases may be wide-spread and carries major implications for design and therapeutic application of kinase inhibitors. DOI: http://dx.doi.org/10.7554/eLife.05434.001


eLife | 2015

Paradoxical resistance of multiple myeloma to proteasome inhibitors by decreased levels of 19S proteasomal subunits

Diego Acosta-Alvear; Min Yi Cho; Thomas Wild; Tonia J. Buchholz; Alana Lerner; Olga Simakova; Jamie Hahn; Neha Korde; Ola Landgren; Irina Maric; Chunaram Choudhary; Peter Walter; Jonathan S. Weissman; Martin Kampmann

Hallmarks of cancer, including rapid growth and aneuploidy, can result in non-oncogene addiction to the proteostasis network that can be exploited clinically. The defining example is the exquisite sensitivity of multiple myeloma (MM) to 20S proteasome inhibitors, such as carfilzomib. However, MM patients invariably acquire resistance to these drugs. Using a next-generation shRNA platform, we found that proteostasis factors, including chaperones and stress-response regulators, controlled the response to carfilzomib. Paradoxically, 19S proteasome regulator knockdown induced resistance to carfilzomib in MM and non-MM cells. 19S subunit knockdown did not affect the activity of the 20S subunits targeted by carfilzomib nor their inhibition by the drug, suggesting an alternative mechanism, such as the selective accumulation of protective factors. In MM patients, lower 19S levels predicted a diminished response to carfilzomib-based therapies. Together, our findings suggest that an understanding of network rewiring can inform development of new combination therapies to overcome drug resistance. DOI: http://dx.doi.org/10.7554/eLife.08153.001


Journal of Clinical Investigation | 2016

Antibody-drug conjugate targeting CD46 eliminates multiple myeloma cells

Daniel W. Sherbenou; Blake T. Aftab; Yang Su; Christopher R. Behrens; Arun P. Wiita; Aaron C Logan; Diego Acosta-Alvear; Byron Hann; Peter Walter; Marc A. Shuman; Xiaobo Wu; John P. Atkinson; Jeffrey L. Wolf; Thomas G. Martin; Bin Liu

Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.

Collaboration


Dive into the Diego Acosta-Alvear's collaboration.

Top Co-Authors

Avatar

Peter Walter

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc A. Shuman

University of California

View shared research outputs
Top Co-Authors

Avatar

Blake T. Aftab

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Min Y. Cho

University of California

View shared research outputs
Top Co-Authors

Avatar

Byron Hann

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge