Network


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

Hotspot


Dive into the research topics where Alemayehu A. Gorfe is active.

Publication


Featured researches published by Alemayehu A. Gorfe.


The EMBO Journal | 2008

A novel switch region regulates H‐ras membrane orientation and signal output

Daniel Abankwa; Michael Hanzal-Bayer; Nicolas Ariotti; Sarah J. Plowman; Alemayehu A. Gorfe; Robert G. Parton; J. Andrew McCammon; John F. Hancock

The plasma membrane nanoscale distribution of H‐ras is regulated by guanine nucleotide binding. To explore the structural basis of H‐ras membrane organization, we combined molecular dynamic simulations and medium‐throughput FRET measurements on live cells. We extracted a set of FRET values, termed a FRET vector, to describe the lateral segregation and orientation of H‐ras with respect to a large set of nanodomain markers. We show that mutation of basic residues in helix α4 or the hypervariable region (HVR) selectively alter the FRET vectors of GTP‐ or GDP‐loaded H‐ras, demonstrating a critical role for these residues in stabilizing GTP‐ or GDP‐H‐ras interactions with the plasma membrane. By a similar analysis, we find that the β2–β3 loop and helix α5 are involved in a novel conformational switch that operates through helix α4 and the HVR to reorient the H‐ras G‐domain with respect to the plasma membrane. Perturbation of these switch elements enhances MAPK activation by stabilizing GTP‐H‐ras in a more productive signalling conformation. The results illustrate how the plasma membrane spatially constrains signalling conformations by acting as a semi‐neutral interaction partner.


Current Opinion in Structural Biology | 2010

Large conformational changes in proteins: Signaling and other functions

Barry J. Grant; Alemayehu A. Gorfe; J. Andrew McCammon

Guanine and adenine nucleotide triphosphatases, such as Ras proteins and protein kinases, undergo large conformational changes upon ligand binding in the course of their functions. New computer simulation methods have combined with experimental studies to deepen our understanding of these phenomena. In particular, a conformational selection picture is emerging, where alterations in the relative populations of pre-existing conformations can best describe the conformational switching activity of these important proteins.


PLOS Computational Biology | 2009

Ras conformational switching: simulating nucleotide-dependent conformational transitions with accelerated molecular dynamics.

Barry J. Grant; Alemayehu A. Gorfe; J. Andrew McCammon

Ras mediates signaling pathways controlling cell proliferation and development by cycling between GTP- and GDP-bound active and inactive conformational states. Understanding the complete reaction path of this conformational change and its intermediary structures is critical to understanding Ras signaling. We characterize nucleotide-dependent conformational transition using multiple-barrier-crossing accelerated molecular dynamics (aMD) simulations. These transitions, achieved for the first time for wild-type Ras, are impossible to observe with classical molecular dynamics (cMD) simulations due to the large energetic barrier between end states. Mapping the reaction path onto a conformer plot describing the distribution of the crystallographic structures enabled identification of highly populated intermediate structures. These structures have unique switch orientations (residues 25–40 and 57–75) intermediate between GTP and GDP states, or distinct loop3 (46–49), loop7 (105–110), and α5 C-terminus (159–166) conformations distal from the nucleotide-binding site. In addition, these barrier-crossing trajectories predict novel nucleotide-dependent correlated motions, including correlations of α2 (residues 66–74) with α3-loop7 (93–110), loop2 (26–37) with loop10 (145–151), and loop3 (46–49) with α5 (152–167). The interconversion between newly identified Ras conformations revealed by this study advances our mechanistic understanding of Ras function. In addition, the pattern of correlated motions provides new evidence for a dynamic linkage between the nucleotide-binding site and the membrane interacting C-terminus critical for the signaling function of Ras. Furthermore, normal mode analysis indicates that the dominant collective motion that occurs during nucleotide-dependent conformational exchange, and captured in aMD (but absent in cMD) simulations, is a low-frequency motion intrinsic to the structure.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Ras membrane orientation and nanodomain localization generate isoform diversity

Daniel Abankwa; Alemayehu A. Gorfe; Kerry L. Inder; John F. Hancock

The structural elements encoding functional diversity among Ras GTPases are poorly defined. The orientation of the G domain of H-ras with respect to the plane of the plasma membrane is recognized by the Ras binding domain of C-Raf, coupling orientation to MAPK activation. We now show that two other proteins, phosphoinositide-3-kinase-α and the structurally unrelated galectin-1, also recognize G-domain orientation. These results rationalize the role of galectin-1 in generating active GTP-H-ras signaling nanoclusters. However, molecular dynamics simulations of K-ras membrane insertion and fluorescence lifetime imaging microscopy (FLIM)-Förster resonance energy transfer (FRET) imaging of the effector interactions of N-Ras, K-Ras, and M-ras suggest that there are two hyperactive, signaling-competent orientations of the Ras G domain. Mutational and functional analyses establish a clear relationship between effector binding and the amphilicities of helix α4 and the C-terminal hypervariable region, thus confirming that these structural elements critically tune the orientation of the Ras G domain. Finally, we show that G-domain orientation and nanoclustering synergize to generate Ras isoform specificity with respect to effector interactions.


Structure | 2008

Mapping the Nucleotide and Isoform-Dependent Structural and Dynamical Features of Ras Proteins

Alemayehu A. Gorfe; Barry J. Grant; J. Andrew McCammon

Ras GTPases are conformational switches controlling cell proliferation, differentiation, and development. Despite their prominent role in many forms of cancer, the mechanism of conformational transition between inactive GDP-bound and active GTP-bound states remains unclear. Here we describe a detailed analysis of available experimental structures and molecular dynamics simulations to quantitatively assess the structural and dynamical features of active and inactive states and their interconversion. We demonstrate that GTP-bound and nucleotide-free G12V H-ras sample a wide region of conformational space, and show that the inactive-to-active transition is a multiphase process defined by the relative rearrangement of the two switches and the orientation of Tyr32. We also modeled and simulated N- and K-ras proteins and found that K-ras is more flexible than N- and H-ras. We identified a number of isoform-specific, long-range side chain interactions that define unique pathways of communication between the nucleotide binding site and the C terminus.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Organization, dynamics, and segregation of Ras nanoclusters in membrane domains

Lorant Janosi; Zhenlong Li; John F. Hancock; Alemayehu A. Gorfe

Recent experiments have shown that membrane-bound Ras proteins form transient, nanoscale signaling platforms that play a crucial role in high-fidelity signal transmission. However, a detailed characterization of these dynamic proteolipid substructures by high-resolution experimental techniques remains elusive. Here we use extensive semiatomic simulations to reveal the molecular basis for the formation and domain-specific distribution of Ras nanoclusters. As model systems, we chose the triply lipidated membrane targeting motif of H-ras (tH) and a large bilayer made up of di16∶0-PC (DPPC), di18∶2-PC (DLiPC), and cholesterol. We found that 4–10 tH molecules assemble into clusters that undergo molecular exchange in the sub-μs to μs time scale, depending on the simulation temperature and hence the stability of lipid domains. Driven by the opposite preference of tH palmitoyls and farnesyl for ordered and disordered membrane domains, clustered tH molecules segregate to the boundary of lipid domains. Additionally, a systematic analysis of depalmitoylated and defarnesylated tH variants allowed us to decipher the role of individual lipid modifications in domain-specific nanocluster localization and thereby explain why homologous Ras isoforms form nonoverlapping nanoclusters. Moreover, the localization of tH nanoclusters at domain boundaries resulted in a significantly lower line tension and increased membrane curvature. Taken together, these results provide a unique mechanistic insight into how protein assembly promoted by lipid-modification modulates bilayer shape to generate functional signaling platforms.


PLOS ONE | 2011

Novel allosteric sites on ras for lead generation

Barry J. Grant; Suryani Lukman; Harrison J. Hocker; Jaqueline Sayyah; Joan Heller Brown; J. Andrew McCammon; Alemayehu A. Gorfe

Aberrant Ras activity is a hallmark of diverse cancers and developmental diseases. Unfortunately, conventional efforts to develop effective small molecule Ras inhibitors have met with limited success. We have developed a novel multi-level computational approach to discover potential inhibitors of previously uncharacterized allosteric sites. Our approach couples bioinformatics analysis, advanced molecular simulations, ensemble docking and initial experimental testing of potential inhibitors. Molecular dynamics simulation highlighted conserved allosteric coupling of the nucleotide-binding switch region with distal regions, including loop 7 and helix 5. Bioinformatics methods identified novel transient small molecule binding pockets close to these regions and in the vicinity of the conformationally responsive switch region. Candidate binders for these pockets were selected through ensemble docking of ZINC and NCI compound libraries. Finally, cell-based assays confirmed our hypothesis that the chosen binders can inhibit the downstream signaling activity of Ras. We thus propose that the predicted allosteric sites are viable targets for the development and optimization of new drugs.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Andrographolide derivatives inhibit guanine nucleotide exchange and abrogate oncogenic Ras function

Harrison J. Hocker; Kwang Jin Cho; Chung Ying K. Chen; Nandini Rambahal; Sreenivasa Rao Sagineedu; Khozirah Shaari; Johnson Stanslas; John F. Hancock; Alemayehu A. Gorfe

Aberrant signaling by oncogenic mutant rat sarcoma (Ras) proteins occurs in ∼15% of all human tumors, yet direct inhibition of Ras by small molecules has remained elusive. Recently, several small-molecule ligands have been discovered that directly bind Ras and inhibit its function by interfering with exchange factor binding. However, it is unclear whether, or how, these ligands could lead to drugs that act against constitutively active oncogenic mutant Ras. Using a dynamics-based pocket identification scheme, ensemble docking, and innovative cell-based assays, here we show that andrographolide (AGP)—a bicyclic diterpenoid lactone isolated from Andrographis paniculata—and its benzylidene derivatives bind to transient pockets on Kirsten-Ras (K-Ras) and inhibit GDP–GTP exchange. As expected for inhibitors of exchange factor binding, AGP derivatives reduced GTP loading of wild-type K-Ras in response to acute EGF stimulation with a concomitant reduction in MAPK activation. Remarkably, however, prolonged treatment with AGP derivatives also reduced GTP loading of, and signal transmission by, oncogenic mutant K-RasG12V. In sum, the combined analysis of our computational and cell biology results show that AGP derivatives directly bind Ras, block GDP–GTP exchange, and inhibit both wild-type and oncogenic K-Ras signaling. Importantly, our findings not only show that nucleotide exchange factors are required for oncogenic Ras signaling but also demonstrate that inhibiting nucleotide exchange is a valid approach to abrogating the function of oncogenic mutant Ras.


PLOS Computational Biology | 2010

The Distinct Conformational Dynamics of K-Ras and H-Ras A59G

Suryani Lukman; Barry J. Grant; Alemayehu A. Gorfe; Guy H. Grant; J. Andrew McCammon

Ras proteins regulate signaling cascades crucial for cell proliferation and differentiation by switching between GTP- and GDP-bound conformations. Distinct Ras isoforms have unique physiological functions with individual isoforms associated with different cancers and developmental diseases. Given the small structural differences among isoforms and mutants, it is currently unclear how these functional differences and aberrant properties arise. Here we investigate whether the subtle differences among isoforms and mutants are associated with detectable dynamical differences. Extensive molecular dynamics simulations reveal that wild-type K-Ras and mutant H-Ras A59G are intrinsically more dynamic than wild-type H-Ras. The crucial switch 1 and switch 2 regions along with loop 3, helix 3, and loop 7 contribute to this enhanced flexibility. Removing the gamma-phosphate of the bound GTP from the structure of A59G led to a spontaneous GTP-to-GDP conformational transition in a 20-ns unbiased simulation. The switch 1 and 2 regions exhibit enhanced flexibility and correlated motion when compared to non-transitioning wild-type H-Ras over a similar timeframe. Correlated motions between loop 3 and helix 5 of wild-type H-Ras are absent in the mutant A59G reflecting the enhanced dynamics of the loop 3 region. Taken together with earlier findings, these results suggest the existence of a lower energetic barrier between GTP and GDP states of the mutant. Molecular dynamics simulations combined with principal component analysis of available Ras crystallographic structures can be used to discriminate ligand- and sequence-based dynamic perturbations with potential functional implications. Furthermore, the identification of specific conformations associated with distinct Ras isoforms and mutants provides useful information for efforts that attempt to selectively interfere with the aberrant functions of these species.


Biophysical Journal | 2010

Conformational Selection in G-Proteins: Lessons from Ras and Rho

Barry J. Grant; J. Andrew McCammon; Alemayehu A. Gorfe

The induced fit model has traditionally been invoked to describe the activating conformational change of the monomeric G-proteins, such as Ras and Rho. With this scheme, the presence or absence of the γ-phosphate of GTP leads to an instantaneous switch in conformation. Here we describe atomistic molecular simulations that demonstrate that both Ras and Rho superfamily members harbor an intrinsic susceptibility to sample multiple conformational states in the absence of nucleotide ligand. By comparing the distribution of conformers in the presence and absence of nucleotide, we show that conformational selection is the dominant mechanism by which Ras and Rho undergo nucleotide-dependent conformational changes. Furthermore, the pattern of correlated motions revealed by these simulations predicts a preserved allosteric coupling of the nucleotide-binding site with the membrane interacting C-terminus in both Rho and Ras.

Collaboration


Dive into the Alemayehu A. Gorfe's collaboration.

Top Co-Authors

Avatar

Priyanka Prakash

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John F. Hancock

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Abdallah Sayyed-Ahmad

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Zhenlong Li

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Xubo Lin

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Ilya Levental

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Lorant Janosi

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Yong Zhou

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge