Network


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

Hotspot


Dive into the research topics where Karthik S. Prabhakara is active.

Publication


Featured researches published by Karthik S. Prabhakara.


Stem Cells Translational Medicine | 2016

Propranolol and Mesenchymal Stromal Cells Combine to Treat Traumatic Brain Injury

Daniel J. Kota; Karthik S. Prabhakara; Alexandra J. van Brummen; Supinder S. Bedi; Hasen Xue; Bryan DiCarlo; Charles S. Cox; Scott D. Olson

More than 6.5 million patients are burdened by the physical, cognitive, and psychosocial deficits associated with traumatic brain injury (TBI) in the U.S. Despite extensive efforts to develop neuroprotective therapies for this devastating disorder, there have been no successful outcomes in human clinical trials to date. Retrospective studies have shown that β‐adrenergic receptor blockers, specifically propranolol, significantly decrease mortality of TBI through mechanisms not yet fully elucidated but are thought to counterbalance a hyperadrenergic state resulting from a TBI. Conversely, cellular therapies have been shown to improve long‐term behavior following TBI, likely by reducing inflammation. Given the nonredundancy in their therapeutic mechanisms, we hypothesized that a combination of acute propranolol followed by mesenchymal stem cells (MSCs) isolated from human bone marrow would have additive effects in treating a rodent model of TBI. We have found that the treatments are well‐tolerated individually and in combination with no adverse events. MSCs decrease BBB permeability at 96 hours after injury, inhibit a significant accumulation of activated microglia/macrophage in the thalamic region of the brain both short and long term, and enhance neurogenesis short term. Propranolol decreases edema and reduces the number of fully activated microglia at 7 days and the number of semiactivated microglia at 120 days. Combinatory treatment improved cognitive and memory functions 120 days following TBI. Therefore, the results here suggest a new, efficacious sequential treatment for TBI may be achieved using the β‐blocker propranolol followed by MSC treatment.


The International Journal of Biochemistry & Cell Biology | 2014

MSCs and hyaluronan: Sticking together for new therapeutic potential?

Daniel J. Kota; Karthik S. Prabhakara; Charles S. Cox; Scott D. Olson

Research involving mesenchymal multipotent/stem/progenitor/stromal/marrow cells (MSCs) have translated to clinical trials at an extraordinary pace. By the time of this review, the public clinical trials database (http://clinicaltrials.gov) has 394 clinical trials listed using MSCs for a very wide range of therapeutic applications. Unexpectedly, the explanation for the increase in clinical trials using MSCs does not lie on a well-defined therapeutic mechanism--dramatic results have been demonstrated in a variety of studies involving different animal models of diseases, often describing discrete therapeutic mechanisms exerted by MSCs. This review will focus on recent data suggesting the involvement of hyaluronic acid (HA) in the beneficial effects of MSCs, evaluate the potential of MSC as modulators of HA and the implications of this modulation for disease therapy.


Stem Cells | 2017

Prostaglandin E2 Indicates Therapeutic Efficacy of Mesenchymal Stem Cells in Experimental Traumatic Brain Injury

Daniel J. Kota; Karthik S. Prabhakara; Naama Toledano‐Furman; Deepa Bhattarai; Qingzheng Chen; Bryan DiCarlo; Philippa Smith; Fabio Triolo; Pamela L. Wenzel; Charles S. Cox; Scott D. Olson

Traumatic brain injury (TBI) is soon predicted to become the third leading cause of death and disability worldwide. After the primary injury, a complex set of secondary injuries develops hours and days later with prolonged neuroinflammation playing a key role. TBI and other inflammatory conditions are currently being treated in preclinical and clinical trials by a number of cellular therapies. Mesenchymal stem cells (MSC) are of great interest due to their widespread usage, safety, and relative ease to isolate and culture. However, there has been a wide range in efficacy reported using MSC clinically and in preclinical models, likely due to differences in cell preparations and a significant amount of donor variability. In this study, we seek to find a correlation between in vitro activity and in vivo efficacy. We designed assays to explore the responsiveness of MSC to immunological cues to address the immunomodulatory properties of MSC, one of their primary modes of therapeutic activity in TBI. Our results showed intrinsic differences in the immunomodulatory capacity of MSC preparations from different bone marrow and amniotic fluid donors. This difference mirrored the therapeutic capacity of the MSC in an experimental model of TBI, an effect confirmed using siRNA knockdown of COX2 followed by overexpressing COX2. Among the immunomodulatory factors assessed, the therapeutic benefit correlated with the secretion of prostaglandin E2 (PGE2) by MSC prior to treatment, suggesting that measurement of PGE2 could be a very useful potency marker to create an index of predicted efficacy for preparations of MSC to treat TBI. Stem Cells 2017;35:1416–1430


Stem Cells | 2018

Inflammation‐Stimulated Mesenchymal Stromal Cell‐Derived Extracellular Vesicles Attenuate Inflammation

Matthew T. Harting; Amit K. Srivastava; Siqin Zhaorigetu; Henry Bair; Karthik S. Prabhakara; Naama E. Toledano Furman; Jody Vykoukal; Katherine A. Ruppert; Charles S. Cox; Scott D. Olson

Extracellular vesicles (EVs) secreted by mesenchymal stromal cells (MSCs) have been proposed to be a key mechanistic link in the therapeutic efficacy of cells in response to cellular injuries through paracrine effects. We hypothesize that inflammatory stimulation of MSCs results in the release of EVs that have greater anti‐inflammatory effects. The present study evaluates the immunomodulatory abilities of EVs derived from inflammation‐stimulated and naive MSCs (MSCEv+ and MSCEv, respectively) isolated using a current Good Manufacturing Practice‐compliant tangential flow filtration system. Detailed characterization of both EVs revealed differences in protein composition, cytokine profiles, and RNA content, despite similarities in size and expression of common surface markers. MSCEv+ further attenuated release of pro‐inflammatory cytokines in vitro when compared to MSCEv, with a distinctly different pattern of EV‐uptake by activated primary leukocyte subpopulations. The efficacy of EVs was partially attributed to COX2/PGE2 expression. The present study demonstrates that inflammatory stimulation of MSCs renders release of EVs that have enhanced anti‐inflammatory properties partially due to COX2/PGE2 pathway alteration. Stem Cells 2018;36:79–90


Cytometry Part A | 2018

OMIP‐041: Optimized multicolor immunofluorescence panel rat microglial staining protocol

Naama E. Toledano Furman; Karthik S. Prabhakara; Supinder S. Bedi; Charles S. Cox; Scott D. Olson

THE common usage of animal models in a variety of preclinical studies is supported by appropriate species-specific antibodies to be utilized in immunohistochemistry (IHC), western blotting, and flow cytometry (FC) assays. Other than the technical advantages (sophisticated surgical manipulations due to their size), modest cost (relative to larger animals), and the standardized results, the similarities in metabolic activity and physiology of neurological disorders to humans make rats appropriate for neurological disease or disorders models (1,2). However, rat-based assays are not as comprehensive or standardized as mouse or human based assays are, partially because there is a shortage in rat-specific antibodies. Rat-specific antibodies are now becoming commercially available, which allows us to set standardize criteria for rat origin cells of interest. As our research focus is in neurotherapy, we are interested specifically in microglial cells, which are the innate immune cells in the brain (3) and spinal cord (4). Microglial cells play a critical role in traumatic brain injuries (TBI) and spinal cord injuries (SCI), and their presence, activation, and effect are highly investigated in those models (4–6). Microglial characterization via FC would save many hours of work as a substitute for IHC analysis, yield unbiased statistics, and overall help research move at a faster pace (7). Here we present multicolor phenotyping panels for assessing microglia derived from rat brain or spinal cord for their activation states, polarization, and number (see Table 1). The microglial cells used are immediately isolated from fresh brain or spinal cord tissues, using a Neural Tissue Dissociation kit, followed by myelin removal and purification using anti-rat CD11b/ c microbeads.


Scientific Reports | 2018

Human Mesenchymal Stromal Cell-Derived Extracellular Vesicles Modify Microglial Response and Improve Clinical Outcomes in Experimental Spinal Cord Injury

Katherine A. Ruppert; Tin T. Nguyen; Karthik S. Prabhakara; Naama E. Toledano Furman; Amit K. Srivastava; Matthew T. Harting; Charles S. Cox; Scott D. Olson

No current clinical intervention can alter the course of acute spinal cord injury (SCI), or appreciably improve neurological outcome. Mesenchymal stromal cells (MSCs) have been shown to modulate the injury sequelae of SCI largely via paracrine effects, although the mechanisms remain incompletely understood. One potential modality is through secretion of extracellular vesicles (EVs). In this study, we investigate whether systemic administration of EVs isolated from human MSCs (MSCEv) has the potential to be efficacious as an alternative to cell-based therapy for SCI. Additionally, we investigate whether EVs isolated from human MSCs stimulated with pro-inflammatory cytokines have enhanced anti-inflammatory effects when administered after SCI. Immunohistochemistry supported the quantitative analysis, demonstrating a diminished inflammatory response with apparent astrocyte and microglia disorganization in cord tissue up to 10 mm caudal to the injury site. Locomotor recovery scores showed significant improvement among animals treated with MSCEv. Significant increases in mechanical sensitivity threshold were observed in animals treated with EVs from either naïve MSC (MSCEvwt) or stimulated MSC (MSCEv+), with a statistically significant increase in threshold for MSCEv+-treated animals when compared to those that received MSCEvwt. In conclusion, these data show that treatment of acute SCI with extracellular vesicles derived from human MSCs attenuates neuroinflammation and improves functional recovery.


Stem Cells Translational Medicine | 2018

Clinical Cellular Therapeutics Accelerate Clot Formation: Cellular Therapeutics Accelerate Clot Formation

Mitchell J. George; Karthik S. Prabhakara; Naama Toledano‐Furman; Yao-Wei Wang; Brijesh S. Gill; Charles E. Wade; Scott D. Olson; Charles S. Cox

Clinical cellular therapeutics (CCTs) have shown preliminary efficacy in reducing inflammation after trauma, preserving cardiac function after myocardial infarction, and improving functional recovery after stroke. However, most clinically available cell lines express tissue factor (TF) which stimulates coagulation. We sought to define the degree of procoagulant activity of CCTs as related to TF expression. CCT samples from bone marrow, adipose, amniotic fluid, umbilical cord, multi‐potent adult progenitor cell donors, and bone marrow mononuclear cells were tested. TF expression and phenotype were quantified using flow cytometry. Procoagulant activity of the CCTs was measured in vitro with thromboelastography and calibrated thrombogram. Fluorescence‐activated cell sorting (FACS) separated samples into high‐ and low‐TF expressing populations to isolate the contribution of TF to coagulation. A TF neutralizing antibody was incubated with samples to demonstrate loss of procoagulant function. All CCTs tested expressed procoagulant activity that correlated with expression of tissue factor. Time to clot and thrombin formation decreased with increasing TF expression. High‐TF expressing cells decreased clotting time more than low‐TF expressing cells when isolated from a single donor using FACS. A TF neutralizing antibody restored clotting time to control values in some, but not all, CCT samples. CCTs demonstrate wide variability in procoagulant activity related to TF expression. Time to clot and thrombin formation decreases as TF load increases and this procoagulant effect is neutralized by a TF blocking antibody. Clinical trials using CCTs are in progress and TF expression may emerge as a safety release criterion. Stem Cells Translational Medicine 2018;7:731–739


Genome Biology | 2018

Alternative splicing links histone modifications to stem cell fate decision

Yungang Xu; Weilling Zhao; Scott D. Olson; Karthik S. Prabhakara; Xiaobo Zhou

BackgroundUnderstanding the embryonic stem cell (ESC) fate decision between self-renewal and proper differentiation is important for developmental biology and regenerative medicine. Attention has focused on mechanisms involving histone modifications, alternative pre-messenger RNA splicing, and cell-cycle progression. However, their intricate interrelations and joint contributions to ESC fate decision remain unclear.ResultsWe analyze the transcriptomes and epigenomes of human ESC and five types of differentiated cells. We identify thousands of alternatively spliced exons and reveal their development and lineage-dependent characterizations. Several histone modifications show dynamic changes in alternatively spliced exons and three are strongly associated with 52.8% of alternative splicing events upon hESC differentiation. The histone modification-associated alternatively spliced genes predominantly function in G2/M phases and ATM/ATR-mediated DNA damage response pathway for cell differentiation, whereas other alternatively spliced genes are enriched in the G1 phase and pathways for self-renewal. These results imply a potential epigenetic mechanism by which some histone modifications contribute to ESC fate decision through the regulation of alternative splicing in specific pathways and cell-cycle genes. Supported by experimental validations and extended datasets from Roadmap/ENCODE projects, we exemplify this mechanism by a cell-cycle-related transcription factor, PBX1, which regulates the pluripotency regulatory network by binding to NANOG. We suggest that the isoform switch from PBX1a to PBX1b links H3K36me3 to hESC fate determination through the PSIP1/SRSF1 adaptor, which results in the exon skipping of PBX1.ConclusionWe reveal the mechanism by which alternative splicing links histone modifications to stem cell fate decision.


Archive | 2018

Rat Microglia Isolation and Characterization Using Multiparametric Panel for Flow Cytometric Analysis

Naama E. Toledano Furman; Karthik S. Prabhakara; Supinder S. Bedi; Charles S. Cox; Scott D. Olson


Molecular Therapy | 2018

Teriflunomide Modulates Vascular Permeability and Microglial Activation after Experimental Traumatic Brain Injury

Karthik S. Prabhakara; Daniel J. Kota; Gregory H. Jones; Amit K. Srivastava; Charles S. Cox; Scott D. Olson

Collaboration


Dive into the Karthik S. Prabhakara's collaboration.

Top Co-Authors

Avatar

Scott D. Olson

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Charles S. Cox

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Daniel J. Kota

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Naama E. Toledano Furman

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Supinder S. Bedi

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Amit K. Srivastava

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Hasen Xue

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Katherine A. Ruppert

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Philippa Smith

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Benjamin M. Aertker

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge