Network


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

Hotspot


Dive into the research topics where Hema Rangaswami is active.

Publication


Featured researches published by Hema Rangaswami.


Journal of Biological Chemistry | 2009

Type II cGMP-dependent Protein Kinase Mediates Osteoblast Mechanotransduction

Hema Rangaswami; Nisha Marathe; Shunhui Zhuang; Yongchang Chen; Jiunn-Chern Yeh; John A. Frangos; Gerry R. Boss; Renate B. Pilz

Continuous bone remodeling in response to mechanical loading is critical for skeletal integrity, and interstitial fluid flow is an important stimulus for osteoblast/osteocyte growth and differentiation. However, the biochemical signals mediating osteoblast anabolic responses to mechanical stimulation are incompletely understood. In primary human osteoblasts and murine MC3T3-E1 cells, we found that fluid shear stress induced rapid expression of c-fos, fra-1, fra-2, and fosB/ΔfosB mRNAs; these genes encode transcriptional regulators that maintain skeletal integrity. Fluid shear stress increased osteoblast nitric oxide (NO) synthesis, leading to activation of cGMP-dependent protein kinase (PKG). Pharmacological inhibition of the NO/cGMP/PKG signaling pathway blocked shear-induced expression of all four fos family genes. Induction of these genes required signaling through MEK/Erk, and Erk activation was NO/cGMP/PKG-dependent. Treating cells with a membrane-permeable cGMP analog partly mimicked the effects of fluid shear stress on Erk activity and fos family gene expression. In cells transfected with small interfering RNAs (siRNA) specific for membrane-bound PKG II, shear- and cGMP-induced Erk activation and fos family gene expression was nearly abolished and could be restored by transducing cells with a virus encoding an siRNA-resistant form of PKG II; in contrast, siRNA-mediated repression of the more abundant cytosolic PKG I isoform was without effect. Thus, we report a novel function for PKG II in osteoblast mechanotransduction, and we propose a model whereby NO/cGMP/PKG II-mediated Erk activation and induction of c-fos, fra-1, fra-2, and fosB/ΔfosB play a key role in the osteoblast anabolic response to mechanical stimulation.


Science Signaling | 2010

Cyclic GMP and Protein Kinase G Control a Src-Containing Mechanosome in Osteoblasts

Hema Rangaswami; Raphaela Schwappacher; Nisha Marathe; Shunhui Zhuang; Darren E. Casteel; Bodo Haas; Yong Chen; Alexander Pfeifer; Hisashi Kato; Sanford J. Shattil; Gerry R. Boss; Renate B. Pilz

Drugs that activate protein kinase G could mimic the bone-building effects of mechanical stimulation. Building Bone The loss of bone density that afflicts individuals with osteoporosis makes them more vulnerable to bone fractures. One way to counteract decreases in bone density is through exercise, which mechanically stimulates bone tissue and initiates proliferation in bone-forming cells. Alternatively, the signaling pathways that mediate this proliferative response could be therapeutically activated to mimic the effects of mechanical stimulation. Nitric oxide (NO) is a second messenger that is produced in response to mechanical stimulation; it triggers production of cyclic GMP (cGMP) and, consequently, activation of protein kinase G (PKG). Rangaswami et al. delineated a pathway in mechanically stimulated osteoblasts whereby activation of PKGII signaling ultimately leads to a proliferative response. Mechanical stimuli triggered the formation of a complex containing PKGII, the tyrosine kinase Src, the phosphatases SHP-1 and SHP-2, and β3 integrin mechanoreceptors. Activation of Src in this complex led to activation of extracellular signal–regulated kinase (ERK), which in turn elicited changes in gene expression that promote proliferation. Thus, PKG-activating drugs could be used to mimic the anabolic effects of mechanical stimulation on bone in the treatment of osteoporosis. The accompanying Perspective by Bidwell and Pavalko describes other examples of signaling pathways that mediate mechanotransduction in bone cells. Mechanical stimulation is crucial for bone growth and remodeling, and fluid shear stress promotes anabolic responses in osteoblasts through multiple second messengers, including nitric oxide (NO). NO triggers production of cyclic guanosine 3′,5′-monophosphate (cGMP), which in turn activates protein kinase G (PKG). We found that the NO-cGMP-PKG signaling pathway activates Src in mechanically stimulated osteoblasts to initiate a proliferative response. PKGII was necessary for Src activation, a process that also required the interaction of Src with β3 integrins and dephosphorylation of Src by a complex containing the phosphatases SHP-1 (Src homology 2 domain–containing tyrosine phosphatase 1) and SHP-2. PKGII directly phosphorylated and stimulated SHP-1 activity, and fluid shear stress triggered the recruitment of PKGII, Src, SHP-1, and SHP-2 to a mechanosome containing β3 integrins. PKGII-null mice showed defective Src and ERK (extracellular signal–regulated kinase) signaling in osteoblasts and decreased ERK-dependent gene expression in bone. Our findings reveal a convergence of NO-cGMP-PKG and integrin signaling and establish a previously unknown mechanism of Src activation. These results support the use of PKG-activating drugs to mimic the anabolic effects of mechanical stimulation of bone in the treatment of osteoporosis.


Journal of Biological Chemistry | 2005

JNK1 Differentially Regulates Osteopontin-induced Nuclear Factorinducing Kinase/MEKK1-dependent Activating Protein-1-mediated Promatrix Metalloproteinase-9 Activation

Hema Rangaswami; Anuradha Bulbule; Gopal C. Kundu

We have recently demonstrated that nuclear factor-inducing kinase (NIK) plays a crucial role in osteopontin (OPN)-induced mitogen-activated protein kinase/IκBα kinase-dependent nuclear factor κB (NFκB)-mediated promatrix metalloproteinase-9 activation (Rangaswami, H., Bulbule, A., and Kundu, G. C. (2004) J. Biol. Chem. 279, 38921-38935). However, the molecular mechanism(s) by which OPN regulates NIK/MEKK1-dependent activating protein-1 (AP-1)-mediated promatrix metalloproteinase-9 activation and whether JNK1 plays any role in regulating both these pathways that control the cell motility are not well defined. Here we report that OPN induces αvβ3 integrin-mediated MEKK1 phosphorylation and MEKK1-dependent JNK1 phosphorylation and activation. Overexpression of NIK enhances OPN-induced c-Jun expression, whereas overexpressed NIK had no role in OPN-induced JNK1 phosphorylation and activation. Sustained activation of JNK1 by overexpression of wild type but not kinase negative MEKK1 resulted in suppression of ERK1/2 activation. But this did not affect the OPN-induced NIK-dependent ERK1/2 activation. OPN stimulated both NIK and MEKK1-dependent c-Jun expression, leading to AP-1 activation, whereas NIK-dependent AP-1 activation is independent of JNK1. OPN also enhanced JNK1-dependent/independent AP-1-mediated urokinase type plasminogen activator (uPA) secretion, uPA-dependent promatrix metalloproteinase-9 (MMP-9) activation, cell motility, and invasion. OPN stimulates tumor growth, and the levels of c-Jun, AP-1, urokinase type plasminogen activator, and MMP-9 were higher in OPN-induced tumor compared with control. To our knowledge this is first report that OPN induces NIK/MEKK1-mediated JNK1-dependent/independent AP-1-mediated pro-MMP-9 activation and regulates the negative crosstalk between NIK/ERK1/2 and MEKK1/JNK1 pathways that ultimately controls the cell motility, invasiveness, and tumor growth.


Journal of Biological Chemistry | 2012

Pro-survival Effects of 17β-Estradiol on Osteocytes Are Mediated by Nitric Oxide/cGMP via Differential Actions of cGMP-dependent Protein Kinases I and II

Nisha Marathe; Hema Rangaswami; Shunhui Zhuang; Gerry R. Boss; Renate B. Pilz

Background: Estrogens prevent bone loss in part by preventing osteocyte apoptosis. Results: Anti-apoptotic effects of 17β-estradiol in osteocytes require NO/cGMP-mediated stimulation of Akt and Akt- and cGMP-dependent protein kinase (PKG)-dependent phosphorylation of BAD. Conclusion: PKG types I and II serve independent anti-apoptotic functions in 17β-estradiol-treated osteocytes, converging on BAD. Significance: These novel mechanisms of 17β-estradiol-mediated bone protection provide a rationale for developing NO/cGMP-based therapies for osteoporosis. Estrogens promote bone health in part by increasing osteocyte survival, an effect that requires activation of the protein kinases Akt and ERK1/2, but the molecular mechanisms involved are only partly understood. Because estrogens increase nitric oxide (NO) synthesis and NO can have anti-apoptotic effects, we examined the role of NO/cGMP signaling in estrogen regulation of osteocyte survival. Etoposide-induced death of MLO-Y4 osteocyte-like cells, assessed by trypan blue staining, caspase-3 cleavage, and TUNEL assays, was completely prevented when cells were pre-treated with 17β-estradiol. This protective effect was mimicked when cells were pre-treated with a membrane-permeable cGMP analog and blocked by pharmacological inhibitors of NO synthase, soluble guanylate cyclase, or cGMP-dependent protein kinases (PKGs), supporting a requirement for NO/cGMP/PKG signaling downstream of 17β-estradiol. siRNA-mediated knockdown and viral reconstitution of individual PKG isoforms demonstrated that the anti-apoptotic effects of estradiol and cGMP were mediated by PKG Iα and PKG II. Akt and ERK1/2 activation by 17β-estradiol required PKG II, and cGMP mimicked the effects of estradiol on Akt and ERK, including induction of ERK nuclear translocation. cGMP induced BAD phosphorylation on several sites, and experiments with phosphorylation-deficient BAD mutants demonstrated that the anti-apoptotic effects of cGMP and 17β-estradiol required BAD phosphorylation on Ser136 and Ser155; these sites were targeted by Akt and PKG I, respectively, and regulate BAD interaction with Bcl-2. In conclusion, 17β-estradiol protects osteocytes against apoptosis by activating the NO/cGMP/PKG cascade; PKG II is required for estradiol-induced activation of ERK and Akt, and PKG Iα contributes to pro-survival signaling by directly phosphorylating BAD.


Journal of Biological Chemistry | 2006

Hypoxia Regulates Cross-talk between Syk and Lck Leading to Breast Cancer Progression and Angiogenesis

Goutam Chakraborty; Hema Rangaswami; Shalini Jain; Gopal C. Kundu

Hypoxia is a key parameter that controls tumor angiogenesis and malignant progression by regulating the expression of several oncogenic molecules. The nonreceptor protein-tyrosine kinases Syk and Lck play crucial roles in the signaling mechanism of various cellular processes. The enhanced expression of Syk in normal breast tissue but not in malignant breast carcinoma has prompted us to investigate its potential role in mammary carcinogenesis. Accordingly, we hypothesized that hypoxia/reoxygenation (H/R) may play an important role in regulating Syk activation, and Lck may be involved in this process. In this study, we have demonstrated that H/R differentially regulates Syk phosphorylation and its subsequent interaction and cross-talk with Lck in MCF-7 cells. Moreover, Syk and Lck play differential roles in regulating Sp1 activation and expressions of melanoma cell adhesion molecule (MelCAM), urokinase-type plasminogen activator (uPA), matrix metalloproteinase-9 (MMP-9), and vascular endothelial growth factor (VEGF) in response to H/R. Overexpression of wild type Syk inhibited the H/R-induced uPA, MMP-9, and VEGF expression but up-regulated MelCAM expression. Our data also indicated that MelCAM acts as a tumor suppressor by negatively regulating H/R-induced uPA secretion and MMP-9 activation. The mice xenograft study showed the cross-talk between Syk and Lck regulated H/R-induced breast tumor progression and further correlated with the expressions of MelCAM, uPA, MMP-9, and VEGF. Human clinical specimen analysis supported the in vitro and in vivo findings. To our knowledge, this is first report that the cross-talk between Syk and Lck regulates H/R-induced breast cancer progression and further suggests that Syk may act as potential therapeutic target for the treatment of breast cancer.


Journal of Biological Chemistry | 2012

Protein Kinase G and Focal Adhesion Kinase Converge on Src/Akt/β-Catenin Signaling Module in Osteoblast Mechanotransduction

Hema Rangaswami; Raphaela Schwappacher; Trish Tran; Geraldine C. Chan; Shunhui Zhuang; Gerry R. Boss; Renate B. Pilz

Background: Fluid shear increases intracellular calcium and NO/cGMP signaling in osteoblasts. Results: Focal adhesion kinase and protein kinase G are independently activated downstream of calcium; both kinases cooperatively activate Src, leading to Akt/GSK3/β-catenin signaling in shear-stressed osteoblasts. Conclusion: Osteoblast mechanotransduction requires cross-talk between FAK and PKG to regulate Akt. Significance: cGMP-elevating agents may prove useful for the treatment of osteoporosis. Mechanical loading of bone induces interstitial fluid flow, leading to fluid shear stress (FSS) of osteoblasts. FSS rapidly increases the intracellular calcium concentration ([Ca2+]) and nitric oxide (NO) synthesis in osteoblasts and activates the protein kinase Akt. Activated Akt stimulates osteoblast proliferation and survival, but the mechanism(s) leading to Akt activation is not well defined. Using pharmacological and genetic approaches in primary human and mouse osteoblasts and mouse MC3T3 osteoblast-like cells, we found that Akt activation by FSS occurred through two parallel pathways; one required calcium stimulation of NO synthase and NO/cGMP/protein kinase G II-dependent activation of Src, and the other required calcium activation of FAK and Src, independent of NO. Both pathways cooperated to increase PI3K-dependent Akt phosphorylation and were necessary for FSS to induce nuclear translocation of β-catenin, c-fos, and cox-2 gene expression and osteoblast proliferation. These data explain how mechanical stimulation of osteoblasts leads to increased signaling through a growth regulatory pathway essential for maintaining skeletal integrity.


Journal of Biological Chemistry | 2012

Rho isoform-specific interaction with IQGAP1 promotes breast cancer cell proliferation and migration.

Darren E. Casteel; Stephanie Turner; Raphaela Schwappacher; Hema Rangaswami; Jacqueline Su-Yuo; Shunhui Zhuang; Gerry R. Boss; Renate B. Pilz

Background: RhoA/C and RhoB have homologous sequences, but opposing functions. Results: IQGAP1 binds prenylated, active RhoA/C, but not RhoB; IQGAP1 increases RhoA/C GTP loading and is required for RhoA/C-induced proliferation and motility of breast cancer cells. Conclusion: IQGAP1 is a regulator and pro-oncogenic effector of RhoA/C. Significance: Disrupting Rho/IQGAP interactions with prenylation inhibitors may be a useful adjunct for breast cancer treatment. We performed a proteomics screen for Rho isoform-specific binding proteins to clarify the tumor-promoting effects of RhoA and C that contrast with the tumor-suppressive effects of RhoB. We found that the IQ-motif-containing GTPase-activating protein IQGAP1 interacts directly with GTP-bound, prenylated RhoA and RhoC, but not with RhoB. Co-immunoprecipitation of IQGAP1 with endogenous RhoA/C was enhanced when RhoA/C were activated by epidermal growth factor (EGF) or transfection of a constitutively active guanine nucleotide exchange factor (GEF). Overexpression of IQGAP1 increased GTP-loading of RhoA/C, while siRNA-mediated depletion of IQGAP1 prevented endogenous RhoA/C activation by growth factors. IQGAP1 knockdown also reduced the amount of GTP bound to GTPase-deficient RhoA/C mutants, suggesting that IQGAP enhances Rho activation by GEF(s) or stabilizes Rho-GTP. IQGAP1 depletion in MDA-MB-231 breast cancer cells blocked EGF- and RhoA-induced stimulation of DNA synthesis. Infecting cells with adenovirus encoding constitutively active RhoAL63 and measuring absolute amounts of RhoA-GTP in infected cells demonstrated that the lack of RhoAL63-induced DNA synthesis in IQGAP1-depleted cells was not due to reduced GTP-bound RhoA. These data suggested that IQGAP1 functions downstream of RhoA. Overexpression of IQGAP1 in MDA-MB-231 cells increased DNA synthesis irrespective of siRNA-mediated RhoA knockdown. Breast cancer cell motility was increased by expressing a constitutively-active RhoCV14 mutant or overexpressing IQGAP1. EGF- or RhoC-induced migration required IQGAP1, but IQGAP1-stimulated migration independently of RhoC, placing IQGAP1 downstream of RhoC. We conclude that IQGAP1 acts both upstream of RhoA/C, regulating their activation state, and downstream of RhoA/C, mediating their effects on breast cancer cell proliferation and migration, respectively.


Journal of Cell Science | 2013

cGMP-dependent protein kinase Iβ regulates breast cancer cell migration and invasion via interaction with the actin/myosin-associated protein caldesmon.

Raphaela Schwappacher; Hema Rangaswami; Jacqueline Su-Yuo; Aaron Hassad; Ryan Spitler; Darren E. Casteel

Summary The two isoforms of type I cGMP-dependent protein kinase (PKGI&agr; and PKGI&bgr;) differ in their first ∼100 amino acids, giving each isoform unique dimerization and autoinhibitory domains. The dimerization domains form coiled-coil structures and serve as platforms for isoform-specific protein–protein interactions. Using the PKGI&bgr; dimerization domain as an affinity probe in a proteomic screen, we identified the actin/myosin-associated protein caldesmon (CaD) as a PKGI&bgr;-specific binding protein. PKGI&bgr; phosphorylated human CaD on serine 12 in vitro and in intact cells. Phosphorylation on serine 12 or mutation of serine 12 to glutamic acid (S12E) reduced the interaction between CaD and myosin IIA. Because CaD inhibits myosin ATPase activity and regulates cell motility, we examined the effects of PKGI&bgr; and CaD on cell migration and invasion. Inhibition of the NO/cGMP/PKG pathway reduced migration and invasion of human breast cancer cells, whereas PKG activation enhanced their motility and invasion. siRNA-mediated knockdown of endogenous CaD had pro-migratory and pro-invasive effects in human breast cancer cells. Reconstituting cells with wild-type CaD slowed migration and invasion; however, CaD containing a phospho-mimetic S12E mutation failed to reverse the pro-migratory and pro-invasive activity of CaD depletion. Our data suggest that PKGI&bgr; enhances breast cancer cell motility and invasive capacity, at least in part, by phosphorylating CaD. These findings identify a pro-migratory and pro-invasive function for PKGI&bgr; in human breast cancer cells, suggesting that PKGI&bgr; is a potential target for breast cancer treatment.


BMC Clinical Pharmacology | 2013

cGMP-dependent protein kinase Iβ phosphorylates and regulates the function of the actin/myosin- associated protein caldesmon

Darren E. Casteel; Raphaela Schwappacher; Hema Rangaswami; Jacqueline Su-Yuo

Background The type I cGMP-dependent protein kinases (PKGIa and PKGIb) are splice variants that differ in their first ~100 amino acids, giving each isoform unique dimerization and autoinhibitory domains. The unique coiled-coil dimerization domains mediate isoform specific proteinprotein interactions, and we have previously identified the amino acids that are important in mediating the interaction between PKGIb and its two known interaction partners, TFII-I and IRAG [1].


BMC Pharmacology | 2011

Src activation by cGMP/PKG II in osteoblasts: characterization of a mechano-sensitive signalling complex

Hema Rangaswami; Raphaela Schwappacher; Nisha Marathe; Darren E. Casteel; Bodo Haas; Alexander Pfeifer; Sanford J. Shattil; Gerry R. Boss; Renate B. Pilz

Author(s): Rangaswami, Hema; Schwappacher, Raphaela; Marathe, Nisha; Casteel, Darren E; Haas, Bodo; Pfeifer, Alexander; Shattil, Sanford; Boss, Gerry; Pilz, Renate B

Collaboration


Dive into the Hema Rangaswami's collaboration.

Top Co-Authors

Avatar

Renate B. Pilz

University of California

View shared research outputs
Top Co-Authors

Avatar

Gerry R. Boss

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shunhui Zhuang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nisha Marathe

University of California

View shared research outputs
Top Co-Authors

Avatar

Gopal C. Kundu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yongchang Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge