Network


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

Hotspot


Dive into the research topics where Chandramohan Kattamuri is active.

Publication


Featured researches published by Chandramohan Kattamuri.


Nature | 2003

Eyes absent represents a class of protein tyrosine phosphatases.

Jayanagendra P. Rayapureddi; Chandramohan Kattamuri; Brian D. Steinmetz; Benjamin J. Frankfort; Edwin Justin Ostrin; Graeme Mardon; Rashmi S. Hegde

The Eyes absent proteins are members of a conserved regulatory network implicated in the development of the eye, muscle, kidney and ear. Mutations in the Eyes absent genes have been associated with several congenital disorders including the multi-organ disease bronchio-oto-renal syndrome, congenital cataracts and late-onset deafness. On the basis of previous analyses it has been shown that Eyes absent is a nuclear transcription factor, acting through interaction with homeodomain-containing Sine oculis (also known as Six) proteins. Here we show that Eyes absent is also a protein tyrosine phosphatase. It does not resemble the classical tyrosine phosphatases that use cysteine as a nucleophile and proceed by means of a thiol-phosphate intermediate. Rather, Eyes absent is the prototype for a class of protein tyrosine phosphatases that use a nucleophilic aspartic acid in a metal-dependent reaction. Furthermore, the phosphatase activity of Eyes absent contributes to its ability to induce eye formation in Drosophila.


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

Growth differentiation factor 9:bone morphogenetic protein 15 heterodimers are potent regulators of ovarian functions

Jia Peng; Qinglei Li; Karen Wigglesworth; Adithya Rangarajan; Chandramohan Kattamuri; Randall T. Peterson; John J. Eppig; Thomas B. Thompson; Martin M. Matzuk

Significance Although genetic studies have uncovered critical functions of GDF9 and BMP15 in female reproduction, many genetic and physiologic data for these ligands remain perplexing. Here we establish that mouse and human GDF9:BMP15 heterodimers are the most biopotent regulators of ovarian granulosa cell functions. Moreover, GDF9:BMP15 heterodimers require a unique signaling complex that includes the type 2 receptor BMPR2, an ALK4/5/7 type 1 kinase receptor, and an ALK6 type 1 co-receptor. GDF9:BMP15 binding to this complex stimulates phosphorylation of SMAD2/3. Our findings explain intraspecies and interspecies functions of these oocyte-synthesized proteins and have key implications for the regulation of female fertility. The TGF-β superfamily is the largest family of secreted proteins in mammals, and members of the TGF-β family are involved in most developmental and physiological processes. Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15), oocyte-secreted paralogs of the TGF-β superfamily, have been shown genetically to control ovarian physiology. Although previous studies found that GDF9 and BMP15 homodimers can modulate ovarian pathways in vitro, the functional species-specific significance of GDF9:BMP15 heterodimers remained unresolved. Therefore, we engineered and produced purified recombinant mouse and human GDF9 and BMP15 homodimers and GDF9:BMP15 heterodimers to compare their molecular characteristics and physiological functions. In mouse granulosa cell and cumulus cell expansion assays, mouse GDF9 and human BMP15 homodimers can up-regulate cumulus expansion-related genes (Ptx3, Has2, and Ptgs2) and promote cumulus expansion in vitro, whereas mouse BMP15 and human GDF9 homodimers are essentially inactive. However, we discovered that mouse GDF9:BMP15 heterodimer is ∼10- to 30-fold more biopotent than mouse GDF9 homodimer, and human GDF9:BMP15 heterodimer is ∼1,000- to 3,000-fold more bioactive than human BMP15 homodimer. We also demonstrate that the heterodimers require the kinase activities of ALK4/5/7 and BMPR2 to activate SMAD2/3 but unexpectedly need ALK6 as a coreceptor in the signaling complex in granulosa cells. Our findings that GDF9:BMP15 heterodimers are the most bioactive ligands in mice and humans compared with homodimers explain many puzzling genetic and physiological data generated during the last two decades and have important implications for improving female fertility in mammals.


Journal of Biological Chemistry | 2008

The Structure of FSTL3·Activin A Complex DIFFERENTIAL BINDING OF N-TERMINAL DOMAINS INFLUENCES FOLLISTATIN-TYPE ANTAGONIST SPECIFICITY

Robin Stamler; Henry T. Keutmann; Yisrael Sidis; Chandramohan Kattamuri; Alan L. Schneyer; Thomas B. Thompson

Transforming growth factor β family ligands are neutralized by a number of structurally divergent antagonists. Follistatin-type antagonists, which include splice variants of follistatin (FS288 and FS315) and follistatin-like 3 (FSTL3), have high affinity for activin A but differ in their affinity for other ligands, particularly bone morphogenetic proteins. To understand the structural basis for ligand specificity within FS-type antagonists, we determined the x-ray structure of activin A in complex with FSTL3 to a resolution of 2.5 Å. Similar to the previously resolved FS·activin A structures, the ligand is encircled by two antagonist molecules blocking all ligand receptor-binding sites. Recently, the significance of the FS N-terminal domain interaction at the ligand type I receptor site has been questioned; however, our data show that for FSTL3, the N-terminal domain forms a more intimate contact with activin A, implying that this interaction is stronger than that for FS. Furthermore, binding studies revealed that replacing the FSTL3 N-terminal domain with the corresponding FS domain considerably lowers activin A affinity. Therefore, both structural and biochemical evidence support a significant interaction of the N-terminal domain of FSTL3 with activin A. In addition, structural comparisons with bone morphogenetic proteins suggest that the interface where the N-terminal domain binds may be the key site for determining FS-type antagonist specificity.


Journal of Biological Chemistry | 2012

Structure of Myostatin·Follistatin-like 3 N-TERMINAL DOMAINS OF FOLLISTATIN-TYPE MOLECULES EXHIBIT ALTERNATE MODES OF BINDING

Jennifer N. Cash; Elizabeth B. Angerman; Chandramohan Kattamuri; Kristof Nolan; Huaying Zhao; Yisrael Sidis; Henry T. Keutmann; Thomas B. Thompson

Background: Myostatin is a strong inhibitor of muscle growth and a therapeutic target for the treatment of muscle wasting. Results: Follistatin-like 3, a myostatin inhibitor, interacts uniquely with myostatin as compared with other ligands through its N-terminal domain. Conclusion: The N-terminal domains of follistatin-type molecules may be specificity determinants in ligand binding. Significance: Follistatin-type molecules form unique, specific interactions with different TGF-β family ligands. TGF-β family ligands are involved in a variety of critical physiological processes. For instance, the TGF-β ligand myostatin is a staunch negative regulator of muscle growth and a therapeutic target for muscle-wasting disorders. Therefore, it is important to understand the molecular mechanisms of TGF-β family regulation. One form of regulation is through inhibition by extracellular antagonists such as the follistatin (Fst)-type proteins. Myostatin is tightly controlled by Fst-like 3 (Fstl3), which is the only Fst-type molecule that has been identified in the serum bound to myostatin. Here, we present the crystal structure of myostatin in complex with Fstl3. The structure reveals that the N-terminal domain (ND) of Fstl3 interacts uniquely with myostatin as compared with activin A, because it utilizes different surfaces on the ligand. This results in conformational differences in the ND of Fstl3 that alter its position in the type I receptor-binding site of the ligand. We also show that single point mutations in the ND of Fstl3 are detrimental to ligand binding, whereas corresponding mutations in Fst have little effect. Overall, we have shown that the NDs of Fst-type molecules exhibit distinctive modes of ligand binding, which may affect overall affinity of ligand·Fst-type protein complexes.


Journal of Molecular Biology | 2012

Members of the DAN Family Are BMP Antagonists That Form Highly Stable Noncovalent Dimers

Chandramohan Kattamuri; David M. Luedeke; Kristof Nolan; Scott A. Rankin; Kenneth D. Greis; Aaron M. Zorn; Thomas B. Thompson

Signaling of bone morphogenetic protein (BMP) ligands is antagonized by a number of extracellular proteins, including noggin, follistatin and members of the DAN (differential screening selected gene abberative in neuroblastoma) family. Structural studies on the DAN family member sclerostin (a weak BMP antagonist) have previously revealed that the protein is monomeric and consists of an eight-membered cystine knot motif with a fold similar to transforming growth factor-β ligands. In contrast to sclerostin, certain DAN family antagonists, including protein related to DAN and cerberus (PRDC), have an unpaired cysteine that is thought to function in covalent dimer assembly (analogous to transforming growth factor-β ligands). Through a combination of biophysical and biochemical studies, we determined that PRDC forms biologically active dimers that potently inhibit BMP ligands. Furthermore, we showed that PRDC dimers, surprisingly, are not covalently linked, as mutation of the unpaired cysteine does not inhibit dimer formation or biological activity. We further demonstrated that the noncovalent PRDC dimers are highly stable under both denaturing and reducing conditions. This study was extended to the founding family member DAN, which also forms noncovalent dimers that are highly stable. These results demonstrate that certain DAN family members can form both monomers and noncovalent dimers, implying that biological activity of DAN family members might be linked to their oligomeric state.


Journal of Biological Chemistry | 2015

Structure of Neuroblastoma Suppressor of Tumorigenicity 1 (NBL1) INSIGHTS FOR THE FUNCTIONAL VARIABILITY ACROSS BONE MORPHOGENETIC PROTEIN (BMP) ANTAGONISTS

Kristof Nolan; Chandramohan Kattamuri; David M. Luedeke; Elizabeth B. Angerman; Scott A. Rankin; Mariana L. Stevens; Aaron M. Zorn; Thomas B. Thompson

Background: NBL1 is a moderate antagonist important for modulating bone morphogenetic protein (BMP) signaling in vivo. Results: Using x-ray crystallography and mutagenesis, regions important for BMP inhibition within NBL1 were identified. Conclusion: Modifications to the BMP binding epitope of NBL1 account for differences in its anti-BMP activity. Significance: This suggests that DAN proteins can be modified to be more effective antagonists for therapeutic purposes. Bone morphogenetic proteins (BMPs) are antagonized through the action of numerous extracellular protein antagonists, including members from the differential screening-selected gene aberrative in neuroblastoma (DAN) family. In vivo, misregulation of the balance between BMP signaling and DAN inhibition can lead to numerous disease states, including cancer, kidney nephropathy, and pulmonary arterial hypertension. Despite this importance, very little information is available describing how DAN family proteins effectively inhibit BMP ligands. Furthermore, our understanding for how differences in individual DAN family members arise, including affinity and specificity, remains underdeveloped. Here, we present the structure of the founding member of the DAN family, neuroblastoma suppressor of tumorigenicity 1 (NBL1). Comparing NBL1 to the structure of protein related to Dan and Cerberus (PRDC), a more potent BMP antagonist within the DAN family, a number of differences were identified. Through a mutagenesis-based approach, we were able to correlate the BMP binding epitope in NBL1 with that in PRDC, where introduction of specific PRDC amino acids in NBL1 (A58F and S67Y) correlated with a gain-of-function inhibition toward BMP2 and BMP7, but not GDF5. Although NBL1S67Y was able to antagonize BMP7 as effectively as PRDC, NBL1S67Y was still 32-fold weaker than PRDC against BMP2. Taken together, this data suggests that alterations in the BMP binding epitope can partially account for differences in the potency of BMP inhibition within the DAN family.


Journal of Biological Chemistry | 2015

Alternative Binding Modes Identified for Growth and Differentiation Factor-associated Serum Protein (GASP) Family Antagonism of Myostatin

Ryan G. Walker; Elizabeth B. Angerman; Chandramohan Kattamuri; Yun Sil Lee; Se-Jin Lee; Thomas B. Thompson

Background: GASP-1 and GASP-2 are highly specific antagonists for the TGF-β ligand myostatin, a negative regulator of muscle growth. Results: GASP-1 and GASP-2 form asymmetric and symmetric complexes with myostatin, respectively. Conclusion: Despite the different binding modes, the GASP proteins retain a high specificity for myostatin. Significance: Inhibition of myostatin can be achieved using different binding modes and may facilitate future development of novel anti-myostatin therapeutics. Myostatin, a member of the TGF-β family of ligands, is a strong negative regulator of muscle growth. As such, it is a prime therapeutic target for muscle wasting disorders. Similar to other TGF-β family ligands, myostatin is neutralized by binding one of a number of structurally diverse antagonists. Included are the antagonists GASP-1 and GASP-2, which are unique in that they specifically antagonize myostatin. However, little is known from a structural standpoint describing the interactions of GASP antagonists with myostatin. Here, we present the First low resolution solution structure of myostatin-free and myostatin-bound states of GASP-1 and GASP-2. Our studies have revealed GASP-1, which is 100 times more potent than GASP-2, preferentially binds myostatin in an asymmetrical 1:1 complex, whereas GASP-2 binds in a symmetrical 2:1 complex. Additionally, C-terminal truncations of GASP-1 result in less potent myostatin inhibitors that form a 2:1 complex, suggesting that the C-terminal domains of GASP-1 are the primary mediators for asymmetric complex formation. Overall, this study provides a new perspective on TGF-β antagonism, where closely related antagonists can utilize different ligand-binding strategies.


Protein Expression and Purification | 2012

Expression and Purification of Recombinant Protein Related to DAN and Cerberus (PRDC)

Chandramohan Kattamuri; David M. Luedeke; Thomas B. Thompson

Bone morphogenetic proteins (BMPs) are secreted protein ligands that control numerous biological processes, such as cell differentiation and cell proliferation. Ligands are regulated by a large number of structurally diverse extracellular antagonists. PRDC or protein related to DAN and cerberus is a BMP antagonist of the DAN family, which is defined by a conserved pattern of cysteine residues that form a ring structure. Here we present the expression and purification of recombinant mouse PRDC (mPRDC) from bacterial (Escherichia coli) inclusion bodies through oxidative refolding. Functional mPRDC was isolated from a nonfunctional component through reverse phase chromatography and shown to inhibit BMP2 and BMP4 in a cell-based luciferase reporter assay. Recombinant mPRDC also bound directly to BMP2, BMP4 and BMP7, but not activin A. Furthermore, circular dichroism indicated that mPRDC is folded and contains a higher than anticipated helical content for a DAN family member protein.


Science Signaling | 2016

MuSK is a BMP co-receptor that shapes BMP responses and calcium signaling in muscle cells

Atilgan Yilmaz; Chandramohan Kattamuri; Rana N. Ozdeslik; Carolyn Schmiedel; Sarah Mentzer; Christoph Schorl; Elena Oancea; Thomas B. Thompson; Justin R. Fallon

In addition to organizing the neuromuscular junction, the receptor tyrosine kinase MuSK is also a co-receptor for BMPs in developing muscle. Muscles need MuSK twice Muscle-specific kinase (MuSK) is a receptor tyrosine kinase that is required for the formation and maintenance of neuromuscular junctions. Yilmaz et al. found that MuSK also functions as a co-receptor for bone morphogenetic proteins (BMPs) in myoblasts and in myotubes. MuSK bound to BMPs and BMP receptors in vitro and promoted signaling by BMPs in muscle cells. MuSK promoted the expression of distinct sets of BMP-induced transcripts in myoblasts and myotubes in a manner that was independent of its kinase activity. One of the transcripts stimulated by MuSK-BMP signaling in myoblasts was required for the ability of BMP4 to inhibit intracellular calcium release in response to activation of muscarinic acetylcholine receptors. Thus, in addition to playing a critical role in organizing the neuromuscular junction, MuSK also acts as a BMP co-receptor in developing muscles. Bone morphogenetic proteins (BMPs) function in most tissues but have cell type–specific effects. Given the relatively small number of BMP receptors, this exquisite signaling specificity requires additional molecules to regulate this pathway’s output. The receptor tyrosine kinase MuSK (muscle-specific kinase) is critical for neuromuscular junction formation and maintenance. Here, we show that MuSK also promotes BMP signaling in muscle cells. MuSK bound to BMP4 and related BMPs with low nanomolar affinity in vitro and to the type I BMP receptors ALK3 and ALK6 in a ligand-independent manner both in vitro and in cultured myotubes. High-affinity binding to BMPs required the third, alternatively spliced MuSK immunoglobulin-like domain. In myoblasts, endogenous MuSK promoted BMP4-dependent phosphorylation of SMADs and transcription of Id1, which encodes a transcription factor involved in muscle differentiation. Gene expression profiling showed that MuSK was required for the BMP4-induced expression of a subset of genes in myoblasts, including regulator of G protein signaling 4 (Rgs4). In myotubes, MuSK enhanced the BMP4-induced expression of a distinct set of genes, including transcripts characteristic of slow muscle. MuSK-mediated stimulation of BMP signaling required type I BMP receptor activity but was independent of MuSK tyrosine kinase activity. MuSK-dependent expression of Rgs4 resulted in the inhibition of Ca2+ signaling induced by the muscarinic acetylcholine receptor in myoblasts. These findings establish that MuSK has dual roles in muscle cells, acting both as a tyrosine kinase–dependent synaptic organizing molecule and as a BMP co-receptor that shapes BMP transcriptional output and cholinergic signaling.


Biochemical Journal | 2017

Analysis and identification of the Grem2 heparin/heparan sulfate-binding motif

Chandramohan Kattamuri; Kristof Nolan; Thomas B. Thompson

Bone morphogenetic proteins (BMPs) are regulated by extracellular antagonists of the DAN (differential screening-selected gene aberrative in neuroblastoma) family. Similar to the BMP ligands, certain DAN family members have been shown to interact with heparin and heparan sulfate (HS). Structural studies of DAN family members Gremlin-1 and Gremlin-2 (Grem2) have revealed a dimeric growth factor-like fold where a series of lysine residues cluster along one face of the protein. In the present study, we used mutagenesis, heparin-binding measurements, and cell surface-binding analysis to identify lysine residues that are important for heparin/HS binding in Grem2. We determined that residues involved in heparin/HS binding, while not necessary for BMP antagonism, merge with the heparin/HS-binding epitope of BMP2. Furthermore, the Grem2-BMP2 complex has higher affinity for heparin than the individual proteins and this affinity is not abrogated when the heparin/HS-binding epitope of Grem2 is attenuated. Overall, the present study shows that the Grem2 heparin/HS and BMP-binding epitopes are unique and independent, where, interestingly, the Grem2-BMP2 complex exhibits a significant increase in binding affinity toward heparin moieties that appear to be partially independent of the Grem2 heparin/HS-binding epitope.

Collaboration


Dive into the Chandramohan Kattamuri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristof Nolan

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Aaron M. Zorn

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rashmi S. Hegde

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Scott A. Rankin

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jayanagendra P. Rayapureddi

Cincinnati Children's Hospital Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge