Network


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

Hotspot


Dive into the research topics where Sharath P. Sasi is active.

Publication


Featured researches published by Sharath P. Sasi.


Oncogene | 2012

Breaking the 'harmony' of TNF-α signaling for cancer treatment.

Sharath P. Sasi; Xinhua Yan; Heiko Enderling; Daniel Park; Hui-ya Gilbert; Cindy Curry; Christina Coleman; Lynn Hlatky; Gangjian Qin; Raj Kishore; David A. Goukassian

Tumor necrosis factor-alpha (TNF-α) binds to two distinct receptors, TNFR1/p55 and TNFR2/p75. TNF-α is implicated in the processes of tumor growth, survival, differentiation, invasion, metastases, secretion of cytokines and pro-angiogenic factors. We have shown that TNFR2/p75 signaling promotes ischemia-induced angiogenesis via modulation of several angiogenic growth factors. We hypothesized that TNFR2/p75 may promote tumor growth and angiogenesis. Growth of mouse Lewis lung carcinoma (LLC1) and/or mouse melanoma B16 cell was evaluated in wild type (WT), p75 knockout (KO) and double p55KO/p75KO mouse tumor xenograft models. Compared with WT and p55KO/p75KO mice, growth of tumors in p75KO mice was significantly decreased (twofold) in both LLC and B16 tumors. Tumor growth inhibition was correlated with decreases in vascular endothelial growth factor (VEGF) expression and capillary density, as well as bone marrow-derived endothelial progenitor cells incorporation into the functional capillary network, and an increase in apoptotic cells in LLC xenografts. Gene array analysis of tumor tissues showed a decrease in gene expression in pathways that promote tumor angiogenesis and cell survival. Blocking p75 by short-hairpin RNA in cultured LLCs led to increases in TNF-mediated apoptosis, as well as decreases in the constitutive and TNF-mediated expression of angiogenic growth factors (VEGF, HGF, PLGF), and SDF-1α receptor CXCR4. In summary, p75 is essential for tumor angiogenesis and survival in highly vascularized murine lung tumor xenografts. Blocking p75 expression may lead to tumor regression. This may represent new and effective therapy against lung neoplasms and potentially tumors of other origin.


PLOS ONE | 2014

Cardiovascular Risks Associated with Low Dose Ionizing Particle Radiation

Xinhua Yan; Sharath P. Sasi; Hannah Gee; Juyong Lee; Yongyao Yang; Raman Mehrzad; Jillian Onufrak; Jin Song; Heiko Enderling; Akhil Agarwal; Layla Rahimi; James P. Morgan; Paul F. Wilson; Joseph P. Carrozza; Kenneth Walsh; Raj Kishore; David A. Goukassian

Previous epidemiologic data demonstrate that cardiovascular (CV) morbidity and mortality may occur decades after ionizing radiation exposure. With increased use of proton and carbon ion radiotherapy and concerns about space radiation exposures to astronauts on future long-duration exploration-type missions, the long-term effects and risks of low-dose charged particle irradiation on the CV system must be better appreciated. Here we report on the long-term effects of whole-body proton (1H; 0.5 Gy, 1 GeV) and iron ion (56Fe; 0.15 Gy, 1GeV/nucleon) irradiation with and without an acute myocardial ischemia (AMI) event in mice. We show that cardiac function of proton-irradiated mice initially improves at 1 month but declines by 10 months post-irradiation. In AMI-induced mice, prior proton irradiation improved cardiac function restoration and enhanced cardiac remodeling. This was associated with increased pro-survival gene expression in cardiac tissues. In contrast, cardiac function was significantly declined in 56Fe ion-irradiated mice at 1 and 3 months but recovered at 10 months. In addition, 56Fe ion-irradiation led to poorer cardiac function and more adverse remodeling in AMI-induced mice, and was associated with decreased angiogenesis and pro-survival factors in cardiac tissues at any time point examined up to 10 months. This is the first study reporting CV effects following low dose proton and iron ion irradiation during normal aging and post-AMI. Understanding the biological effects of charged particle radiation qualities on the CV system is necessary both for the mitigation of space exploration CV risks and for understanding of long-term CV effects following charged particle radiotherapy.


Journal of Oncology | 2010

Inhibition of Melanoma Angiogenesis by Telomere Homolog Oligonucleotides

Christina Coleman; Danielle Levine; Raj Kishore; Gangjian Qin; Tina Thorne; Erin Lambers; Sharath P. Sasi; Mina Yaar; Barbara A. Gilchrest; David A. Goukassian

Telomere homolog oligonucleotides (T-oligos) activate an innate telomere-based program that leads to multiple anticancer effects. T-oligos act at telomeres to initiate signaling through the Werner protein and ATM kinase. We wanted to determine if T-oligos have antiangiogenic effects. We found that T-oligo-treated human melanoma (MM-AN) cells had decreased expression of vascular endothelial growth factor (VEGF), VEGF receptor 2, angiopoeitin-1 and -2 and decreased VEGF secretion. T-oligos activated the transcription factor E2F1 and inhibited the activity of the angiogenic transcription factor, HIF-1α. T-oligos inhibited EC tubulogenesis and total tumor microvascular density matrix invasion by MM-AN cells and ECs in vitro. In melanoma SCID xenografts, two systemic T-oligo injections decreased by 60% (P < .004) total tumor microvascular density and the functional vessels density by 80% (P < .002). These findings suggest that restriction of tumor angiogenesis is among the hosts innate telomere-based anticancer responses and provide further evidence that T-oligos may offer a powerful new approach for melanoma treatment.


American Journal of Physiology-heart and Circulatory Physiology | 2015

Low-dose radiation affects cardiac physiology: gene networks and molecular signaling in cardiomyocytes

Matthew A. Coleman; Sharath P. Sasi; Jillian Onufrak; Mohan Natarajan; Krishnan Manickam; John Schwab; Sujatha Muralidharan; Leif E. Peterson; Yuriy O. Alekseyev; Xinhua Yan; David A. Goukassian

There are 160,000 cancer patients worldwide treated with particle radiotherapy (RT). With the advent of proton, and high (H) charge (Z) and energy (E) HZE ionizing particle RT, the cardiovascular diseases risk estimates are uncertain. In addition, future deep space exploratory-type missions will expose humans to unknown but low doses of particle irradiation (IR). We examined molecular responses using transcriptome profiling in left ventricular murine cardiomyocytes isolated from mice that were exposed to 90 cGy, 1 GeV proton ((1)H) and 15 cGy, 1 GeV/nucleon iron ((56)Fe) over 28 days after exposure. Unsupervised clustering analysis of gene expression segregated samples according to the IR response and time after exposure, with (56)Fe-IR showing the greatest level of gene modulation. (1)H-IR showed little differential transcript modulation. Network analysis categorized the major differentially expressed genes into cell cycle, oxidative responses, and transcriptional regulation functional groups. Transcriptional networks identified key nodes regulating expression. Validation of the signal transduction network by protein analysis and gel shift assay showed that particle IR clearly regulates a long-lived signaling mechanism for ERK1/2, p38 MAPK signaling and identified NFATc4, GATA4, STAT3, and NF-κB as regulators of the response at specific time points. These data suggest that the molecular responses and gene expression to (56)Fe-IR in cardiomyocytes are unique and long-lasting. Our study may have significant implications for the efforts of National Aeronautics and Space Administration to develop heart disease risk estimates for astronauts and for patients receiving conventional and particle RT via identification of specific HZE-IR molecular markers.


Journal of Biological Chemistry | 2015

TNF-TNFR2/p75 signaling inhibits early and increases delayed nontargeted effects in bone marrow-derived endothelial progenitor cells

Sharath P. Sasi; Jin Song; Daniel Park; Heiko Enderling; J. Tyson McDonald; Hannah Gee; Brittany Garrity; Alexander Shtifman; Xinhua Yan; Kenneth Walsh; Mohan Natarajan; Raj Kishore; David A. Goukassian

Background: Ionizing radiation can induce DNA damage in nonirradiated (N-IR) cells via nontargeted effects (NTE). Results: TNF-α and IL-1α mediate NTE in N-IR bone marrow-derived EPCs, and neutralizing TNF-α diminishes NTE in WT and p55 knock-out BM-EPCs. Conclusion: TNF-TNFR2/p75 signaling alters accumulation of inflammatory cytokines that attenuate NTE in N-IR EPCs. Significance: TNFR2/p75 may represent a gene target for mitigation of delayed RBR in BM-EPCs. TNF-α, a pro-inflammatory cytokine, is highly expressed after being irradiated (IR) and is implicated in mediating radiobiological bystander responses (RBRs). Little is known about specific TNF receptors in regulating TNF-induced RBR in bone marrow-derived endothelial progenitor cells (BM-EPCs). Full body γ-IR WT BM-EPCs showed a biphasic response: slow decay of p-H2AX foci during the initial 24 h and increase between 24 h and 7 days post-IR, indicating a significant RBR in BM-EPCs in vivo. Individual TNF receptor (TNFR) signaling in RBR was evaluated in BM-EPCs from WT, TNFR1/p55KO, and TNFR2/p75KO mice, in vitro. Compared with WT, early RBR (1–5 h) were inhibited in p55KO and p75KO EPCs, whereas delayed RBR (3–5 days) were amplified in p55KO EPCs, suggesting a possible role for TNFR2/p75 signaling in delayed RBR. Neutralizing TNF in γ-IR conditioned media (CM) of WT and p55KO BM-EPCs largely abolished RBR in both cell types. ELISA protein profiling of WT and p55KO EPC γ-IR-CM over 5 days showed significant increases in several pro-inflammatory cytokines, including TNF-α, IL-1α (Interleukin-1 alpha), RANTES (regulated on activation, normal T cell expressed and secreted), and MCP-1. In vitro treatments with murine recombinant (rm) TNF-α and rmIL-1α, but not rmMCP-1 or rmRANTES, increased the formation of p-H2AX foci in nonirradiated p55KO EPCs. We conclude that TNF-TNFR2 signaling may induce RBR in naïve BM-EPCs and that blocking TNF-TNFR2 signaling may prevent delayed RBR in BM-EPCs, conceivably, in bone marrow milieu in general.


Radiation Research | 2013

Divergent Modification of Low-Dose 56Fe-Particle and Proton Radiation on Skeletal Muscle

Alexander Shtifman; Matthew J. Pezone; Sharath P. Sasi; Akhil Agarwal; Hannah Gee; Jin Song; Aleksandr Perepletchikov; Xinhua Yan; Raj Kishore; David A. Goukassian

It is unknown whether loss of skeletal muscle mass and function experienced by astronauts during space flight could be augmented by ionizing radiation (IR), such as low-dose high-charge and energy (HZE) particles or low-dose high-energy proton radiation. In the current study adult mice were irradiated whole-body with either a single dose of 15 cGy of 1 GeV/n 56Fe-particle or with a 90 cGy proton of 1 GeV/n proton particles. Both ionizing radiation types caused alterations in the skeletal muscle cytoplasmic Ca2+ ([Ca2+]i) homeostasis. 56Fe-particle irradiation also caused a reduction of depolarization-evoked Ca2+ release from the sarcoplasmic reticulum (SR). The increase in the [Ca2+]i was detected as early as 24 h after 56Fe-particle irradiation, while effects of proton irradiation were only evident at 72 h. In both instances [Ca2+]i returned to baseline at day 7 after irradiation. All 56Fe-particle irradiated samples revealed a significant number of centrally localized nuclei, a histologic manifestation of regenerating muscle, 7 days after irradiation. Neither unirradiated control or proton-irradiated samples exhibited such a phenotype. Protein analysis revealed significant increase in the phosphorylation of Akt, Erk1/2 and rpS6k on day 7 in 56Fe-particle irradiated skeletal muscle, but not proton or unirradiated skeletal muscle, suggesting activation of pro-survival signaling. Our findings suggest that a single low-dose 56Fe-particle or proton exposure is sufficient to affect Ca2+ homeostasis in skeletal muscle. However, only 56Fe-particle irradiation led to the appearance of central nuclei and activation of pro-survival pathways, suggesting an ongoing muscle damage/recovery process.


Frontiers in Oncology | 2015

Ionizing Particle Radiation as a Modulator of Endogenous Bone Marrow Cell Reprogramming: Implications for Hematological Cancers

Sujatha Muralidharan; Sharath P. Sasi; Maria A. Zuriaga; Karen K. Hirschi; Christopher D. Porada; Matthew A. Coleman; Kenneth Walsh; Xinhua Yan; David A. Goukassian

Exposure of individuals to ionizing radiation (IR), as in the case of astronauts exploring space or radiotherapy cancer patients, increases their risk of developing secondary cancers and other health-related problems. Bone marrow (BM), the site in the body where hematopoietic stem cell (HSC) self-renewal and differentiation to mature blood cells occurs, is extremely sensitive to low-dose IR, including irradiation by high-charge and high-energy particles. Low-dose IR induces DNA damage and persistent oxidative stress in the BM hematopoietic cells. Inefficient DNA repair processes in HSC and early hematopoietic progenitors can lead to an accumulation of mutations whereas long-lasting oxidative stress can impair hematopoiesis itself, thereby causing long-term damage to hematopoietic cells in the BM niche. We report here that low-dose 1H- and 56Fe-IR significantly decreased the hematopoietic early and late multipotent progenitor (E- and L-MPP, respectively) cell numbers in mouse BM over a period of up to 10 months after exposure. Both 1H- and 56Fe-IR increased the expression of pluripotent stem cell markers Sox2, Nanog, and Oct4 in L-MPPs and 10 months post-IR exposure. We postulate that low doses of 1H- and 56Fe-IR may induce endogenous cellular reprogramming of BM hematopoietic progenitor cells to assume a more primitive pluripotent phenotype and that IR-induced oxidative DNA damage may lead to mutations in these BM progenitors. This could then be propagated to successive cell lineages. Persistent impairment of BM progenitor cell populations can disrupt hematopoietic homeostasis and lead to hematologic disorders, and these findings warrant further mechanistic studies into the effects of low-dose IR on the functional capacity of BM-derived hematopoietic cells including their self-renewal and pluripotency.


PLOS ONE | 2014

Therapeutic non-toxic doses of TNF induce significant regression in TNFR2-p75 knockdown Lewis lung carcinoma tumor implants.

Sharath P. Sasi; Sanggyu Bae; Jin Song; Aleksandr Perepletchikov; Douglas Schneider; Joseph P. Carrozza; Xinhua Yan; Raj Kishore; Heiko Enderling; David A. Goukassian

Tumor necrosis factor-alpha (TNF) binds to two receptors: TNFR1/p55-cytotoxic and TNFR2/p75-pro-survival. We have shown that tumor growth in p75 knockout (KO) mice was decreased more than 2-fold in Lewis lung carcinoma (LLCs). We hypothesized that selective blocking of TNFR2/p75 LLCs may sensitize them to TNF-induced apoptosis and affect the tumor growth. We implanted intact and p75 knockdown (KD)-LLCs (>90%, using shRNA) into wild type (WT) mice flanks. On day 8 post-inoculation, recombinant murine (rm) TNF-α (12.5 ng/gr of body weight) or saline was injected twice daily for 6 days. Tumor volumes (tV) were measured daily and tumor weights (tW) on day 15, when study was terminated due to large tumors in LLC+TNF group. Tubular bones, spleens and peripheral blood (PB) were examined to determine possible TNF toxicity. There was no significant difference in tV or tW between LLC minus (-) TNF and p75KD/LLC-TNF tumors. Compared to 3 control groups, p75KD/LLC+TNF showed >2-5-fold decreases in tV (p<0.001) and tW (p<0.0001). There was no difference in tV or tW end of study vs. before injections in p75KD/LLC+TNF group. In 3 other groups tV and tW were increased 2.7-4.5-fold (p<0.01, p<0.0002 and p<0.0001). Pathological examination revealed that 1/3 of p75KD/LLC+rmTNF tumors were 100% necrotic, the remaining revealed 40-60% necrosis. No toxicity was detected in bone marrow, spleen and peripheral blood. We concluded that blocking TNFR2/p75 in LLCs combined with intra-tumoral rmTNF injections inhibit LLC tumor growth. This could represent a novel and effective therapy against lung neoplasms and a new paradigm in cancer therapeutics.


Stem Cells International | 2015

Particle Radiation-Induced Nontargeted Effects in Bone-Marrow-Derived Endothelial Progenitor Cells

Sharath P. Sasi; Daniel Park; Sujatha Muralidharan; Justin Wage; Albert Kiladjian; Jillian Onufrak; Heiko Enderling; Xinhua Yan; David A. Goukassian

Bone-marrow- (BM-) derived endothelial progenitor cells (EPCs) are critical for endothelial cell maintenance and repair. During future space exploration missions astronauts will be exposed to space irradiation (IR) composed of a spectrum of low-fluence protons (1H) and high charge and energy (HZE) nuclei (e.g., iron-56Fe) for extended time. How the space-type IR affects BM-EPCs is limited. In media transfer experiments in vitro we studied nontargeted effects induced by 1H- and 56Fe-IR conditioned medium (CM), which showed significant increase in the number of p-H2AX foci in nonirradiated EPCs between 2 and 24 h. A 2–15-fold increase in the levels of various cytokines and chemokines was observed in both types of IR-CM at 24 h. Ex vivo analysis of BM-EPCs from single, low-dose, full-body 1H- and 56Fe-IR mice demonstrated a cyclical (early 5–24 h and delayed 28 days) increase in apoptosis. This early increase in BM-EPC apoptosis may be the effect of direct IR exposure, whereas late increase in apoptosis could be a result of nontargeted effects (NTE) in the cells that were not traversed by IR directly. Identifying the role of specific cytokines responsible for IR-induced NTE and inhibiting such NTE may prevent long-term and cyclical loss of stem and progenitors cells in the BM milieu.


Journal of Radiation Research | 2014

Radiation-associated degenerative cardiovascular risks during normal aging and after adverse CV event 10 months post-initial exposure

Sharath P. Sasi; Xinhua Yan; Juyong Lee; Hamayak Sisakyan; Joseph P. Carrozza; David A. Goukassian

Background: During the future exploration-type space missions, astronauts will be exposed to ionizing radiation (IR) for more than 1–2 years. The effect of cosmic IR during and after space flights on the cardiovascular (CV) system is unknown. Therefore, it is important to evaluate space IR effects on the CV system and determine potential post-mission degenerative excess relative risks (ERR) to the heart as a function of normal aging (IR + AGING model) as well as determine whether space IR may affect the processes of recovery after an adverse CV event (i.e. acute myocardial infarct, AMI) during normal aging (IR + AGING + AMI model). Methods: Nine-month-old C57BL6N male mice were IR once with proton (50 cGy, 1 GeV/n) or (56Fe 15 cGy, 1 GeV/n). IR-induced alterations in cardiac function were assessed by echocardiography (ECHO) and hemodynamic measurements (HEMO). AMI was induced by ligation of left anterior descending (LAD) coronary artery 10 months post-IR. Mice were monitored over 28 days post-AMI. Results: Compared with control, in the IR + AGING study group, left ventricular end-systolic pressure (LVESP) was significantly decreased in both 1H- and 56Fe-IR (P < 0.03, both), suggesting IR-associated decrease in contractile function 10 month post-IRs. However, compared with age-matched control mice (18 months), the LV end-diastolic pressure (LVEDP) was significantly increased (P < 0.05) and minimum LV pressure change (dP/dt min, mmHg/sec) was significantly decreased (P < 0.02) in 1H-IR but not 56Fe-IR mice, suggesting that a single 50 cGy full body 1H-IR decreases considerably the relaxation function of the heart 10 months post-AMI. Of note, an increase in LVEDP and a decrease in dP/dt min are indicators that heart is not pumping blood well and is an early independent prognostic CV risk factor for development of cardiac de-compensation. In all three IR + AGING + AMI study groups, in average, there was 10–15% mortality up to 3 days post-AMI surgery with ∼90% survival rate in all groups 28 days post-AMI. This is rather very good survival rate for 18- to 20-month-old mice after permanent LAD ligation. In the IR + AGING + AMI study group, the most harmful effects on myocardial recovery 10 months post-IR and 28 days post-AMI were observed in the 56Fe-IR group. LVESP was significantly decreased in 56Fe-IR vs control and 1H-IR mice (P < 0.04 and <0.02, respectively). LVEDP was 3-fold higher in 56Fe-IR vs 1H-IR mice (P < 0.004) but was only slightly higher (P = n.s.) compared with control mice. However, dP/dt max and dP/dt min were significantly decreased in 56Fe-IR vs control (P < 0.007 and <0.05, respectively) and 1H-IR mice (P < 0.0004 and <0.0015, respectively), suggesting that 56Fe-AMI hearts developed cardiac de-compensation. Summary: Our data in the IR + AGING study group strongly suggest that 10 months post-IR low-dose high-energy 1H-IR but not low-dose HZE (56Fe) particle IR affects considerably contractile and relaxation functions during normal aging. Conversely, our data in the IR + AGING + AMI study group at 10 months post-IR taken together with our previously reported data for AMI recovery 3 month after a single 50 cGy 1H-IR and 15 cGy 56Fe-IR indicate that 3 months and as long as 10 months after a single full-body IR, the 56Fe-IR is detrimental, whereas 1H-IR does not have negative effects on post-AMI recovery. In fact, single 1H-IR, at this dose, was considerably beneficial for post-AMI recovery at 3 months, as well as at 10 months post-IR. Major conclusions: Our longitudinal 1, 3 and 10 months studies in the IR + AGING and IR + AGING + AMI groups reveal that a single full-body low-dose 1H and HZE particle radiation (56Fe) have long-lasting negative effect on heart homeostasis during normal aging (predominantly 56Fe and at 10 months 1H-IR, as well), and present a significant CV risk for recovery after adverse CV event (exclusively 56Fe-IR, whereas 1H-IR at this dose could beneficial). Further, the divergent effects of low dose 1H-IR vs 56Fe-IR on heart function during normal aging vs after adverse CV event suggest significantly different biological responses responsible for this ion-dependent dichotomy over 10 months post-IR and necessitate further in-depth studies into underlying molecular mechanisms.

Collaboration


Dive into the Sharath P. Sasi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juyong Lee

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

James P. Morgan

Prince Henry's Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge