Network


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

Hotspot


Dive into the research topics where Chad Sanada is active.

Publication


Featured researches published by Chad Sanada.


Experimental Hematology | 2011

Phenotypic correction of hemophilia A in sheep by postnatal intraperitoneal transplantation of FVIII-expressing MSC

Christopher D. Porada; Chad Sanada; Chung-Jung Kuo; Evan Colletti; Walter Mandeville; John Hasenau; Esmail D. Zanjani; Robert Moot; Christopher B. Doering; H. Trent Spencer; Graça Almeida-Porada

We recently re-established a line of sheep that accurately mimics the clinical symptoms and genetics of severe hemophilia A (HA). Here, we tested a novel, nonablative transplantation therapy in two pediatric HA animals. Paternal mesenchymal stem cells (MSC) were transduced with a porcine FVIII-encoding lentivector and transplanted via the intraperitoneal route without preconditioning. At the time of transplantation, these animals had received multiple human FVIII treatments for various spontaneous bleeds and had developed debilitating hemarthroses, which produced severe defects in posture and gait. Transplantation of transduced MSC resolved all existent hemarthroses, and spontaneous bleeds ceased. Damaged joints recovered fully; the animals regained normal posture and gait and resumed normal activity. Despite achieving factor-independence, a sharp rise in pre-existent Bethesda titers occurred following transplantation, decreasing the effectiveness and duration of therapy. Postmortem examination revealed widespread engraftment, with MSC present within the lung, liver, intestine, and thymus, but particularly within joints affected at the time of transplantation, suggesting MSC homed to sites of ongoing injury/inflammation to release FVIII, explaining the dramatic improvement in hemarthrotic joints. In summary, this novel, nonablative MSC transplantation was straightforward, safe, and converted life-threatening, debilitating HA to a moderate phenotype in a large animal model.


Experimental Hematology | 2008

Development and characterization of a novel CD34 monoclonal antibody that identifies sheep hematopoietic stem/progenitor cells

Christopher D. Porada; Duygu Dee Harrison-Findik; Chad Sanada; Vincent Valiente; David S. Thain; Paul J. Simmons; Graça Almeida-Porada; Esmail D. Zanjani

OBJECTIVE We and many others have long used sheep as a predictive model system in which to explore stem cell transplantation. Unfortunately, while numerous markers are available to identify and isolate human hematopoietic stem cells (HSC), no reagents exist that allow HSC/progenitors from sheep to be identified or purified, greatly impeding the application of this well-established large animal model to the study of autologous or allogeneic HSC transplantation. The current studies were undertaken to create a monoclonal antibody to sheep CD34 that would enable isolation and study of sheep HSC/progenitors. MATERIALS AND METHODS A partial cDNA to the extracellular domain of the sheep CD34 antigen was polymerase chain reaction cloned, characterized, and used to genetically immunize mice and create hybridomas. RESULTS The resultant monoclonal antibody to sheep CD34 allows flow cytometric detection of sheep HSC/progenitors present within bone marrow, cord blood, and mobilized peripheral blood. Moreover, this antibody can be used to enrich for HSC/progenitors with enhanced in vitro colony-forming potential, and also identifies endothelial cells in situ within paraffin-embedded tissue sections, similarly to antibodies to human CD34. CONCLUSIONS The availability of this monoclonal antibody recognizing the stem cell antigen CD34 in sheep will greatly facilitate the study of autologous and allogeneic HSC transplantation using this clinically relevant large animal model.


Journal of Cellular Physiology | 2013

Mesenchymal stem cells contribute to endogenous FVIII:c production.

Chad Sanada; Chung-Jung Kuo; Evan Colletti; Melisa Soland; Saloomeh Mokhtari; Mary Ann Knovich; John Owen; Esmail D. Zanjani; Christopher D. Porada; Graça Almeida-Porada

Besides the liver, it has been difficult to identify which organ(s) and/or cellular component(s) contribute significantly to the production of human FVIII:c (FVIII). Thus far, only endothelial cells have been shown to constitute a robust extrahepatic source of FVIII, possibly explaining both the diverse presence of FVIII mRNA in the body, and the observed increase in FVIII levels during liver failure. Here, we investigate whether human mesenchymal stem cells (MSC), ubiquitously present in different organs, could also contribute to FVIII production. MSC isolated from human lung, liver, brain, and bone marrow expressed FVIII message as determined by quantitative‐RT‐PCR. Using an antibody specific for FVIII, confocal microscopy, and umbilical cord‐derived endothelial cells (HUVEC) as a negative control, we demonstrated that, in MSC, FVIII protein was not stored in granules; rather, it localized to the perinuclear region. Furthermore, functional FVIII was detected in MSC supernatants and cell lysates by aPTT and chromogenic assays. These results demonstrate that MSC can contribute at low levels to the functional FVIII pool, and advance the understanding of the physiology of FVIII production and secretion. J. Cell. Physiol.


Frontiers in Bioengineering and Biotechnology | 2016

Fluidic Logic Used in a Systems Approach to Enable Integrated Single-Cell Functional Analysis

Naveen Ramalingam; Brian Fowler; Lukasz Szpankowski; Anne A. Leyrat; Kyle Hukari; Myo Thu Maung; Wiganda Yorza; Michael Norris; Chris Cesar; Joe Shuga; Michael L. Gonzales; Chad Sanada; Xiaohui Wang; Rudy Yeung; Win Hwang; Justin Axsom; Naga Sai Gopi Krishna Devaraju; Ninez Delos Angeles; Cassandra Greene; Ming-Fang Zhou; Eng-Seng Ong; Chang-Chee Poh; Marcos Lam; Henry Choi; Zaw Htoo; Leo Lee; Chee-Sing Chin; Zhong-Wei Shen; Chong T. Lu; Ilona Holcomb

The study of single cells has evolved over the past several years to include expression and genomic analysis of an increasing number of single cells. Several studies have demonstrated wide spread variation and heterogeneity within cell populations of similar phenotype. While the characterization of these populations will likely set the foundation for our understanding of genomic- and expression-based diversity, it will not be able to link the functional differences of a single cell to its underlying genomic structure and activity. Currently, it is difficult to perturb single cells in a controlled environment, monitor and measure the response due to perturbation, and link these response measurements to downstream genomic and transcriptomic analysis. In order to address this challenge, we developed a platform to integrate and miniaturize many of the experimental steps required to study single-cell function. The heart of this platform is an elastomer-based integrated fluidic circuit that uses fluidic logic to select and sequester specific single cells based on a phenotypic trait for downstream experimentation. Experiments with sequestered cells that have been performed include on-chip culture, exposure to various stimulants, and post-exposure image-based response analysis, followed by preparation of the mRNA transcriptome for massively parallel sequencing analysis. The flexible system embodies experimental design and execution that enable routine functional studies of single cells.


Cancer Research | 2017

Abstract 2923: Label-free enrichment and integrated full-length mRNA transcriptome analysis of single live circulating tumor cells from breast cancer patients

Naveen Ramalingam; Yi Fang Lee; Lukasz Szpankowski; Anne A. Leyrat; Brian Fowler; Jovina Tan; Chong Tracy Lu; Ninez Delos Angeles; Chad Sanada; Cassandra Greene; Kyle W. Hukari; Andrew Wu; Yoon-Sim Yap; Jay A.A. West; Ali Asgar S. Bhagat

Background Label-free methods for isolating circulating tumor cells (CTCs) are attractive because they provide an opportunity to analyze a larger set of CTCs that may otherwise be missed due to variable or no expression of protein (label) markers. Understanding genetic and functional heterogeneity in CTCs allows us to gain insight into the mechanisms underscoring metastasis, drug resistance, and tumor aggressiveness. Currently, a simple workflow for isolation and molecular characterization of single CTCs by mRNA sequencing is lacking. In order to address this challenge, we developed a label-free workflow to isolate CTCs from breast cancer patients for full-length mRNA sequencing analysis by integrating the ClearCell® FX System with the Polaris™ system. The ClearCell FX system processes blood samples from cancer patients and enriches for CTCs in a label-free antibody-independent manner. The low level of nonspecifically isolated white blood cells from ClearCell FX is further depleted on the Polaris system by negative enrichment of viable CTCs. This unique integration of systems will enable researchers to perturb single CTCs in a controlled environment, monitor and measure the response due to perturbation, and link these response measurements to downstream genomic and transcriptomic analysis. Method and Results CTCs from 7.5 mL of peripheral blood sample from breast cancer patients were enriched using ClearCell FX. To differentiate larger blood cells from putative CTCs, we stained the enriched cells with Alexa Fluor® 647-conjugated CD45 and CD31 to identify leukocytes and endothelial cells, respectively. Calcein AM (live cell marker) and CellTracker™ Orange (universal cell marker) were added to identify live cells. Single CTCs were selected on Polaris (Fluidigm) system, lysed and reverse-transcribed, and cDNA were preamplified on the Polaris integrated fluidic circuit (IFC). Sequencing libraries were generated using the Nextera® kit and sequenced on Illumina® MiSeq™ and NextSeq™ systems. We successfully processed blood samples from four patients. Sequenced data showed high-quality metrics, with read depth of up to 2.5 million reads (MiSeq) or 60 million reads (NextSeq), with a low percentage of mapped reads to ribosomal RNA and mitochondrial RNA. Unsupervised hierarchical clustering of gene expression data showed clustering by patient, but considerable heterogeneity was also observed among the CTCs from the same patient. We will provide insights into full-length mRNA transcriptome of single CTCs from triple negative breast cancer patient. Conclusion We present the feasibility of integrating two microfluidics platforms to capture single CTCs for transcriptome and functional study. Our data suggests that the heterogeneity of tumor sample and characterization of metastatic processes can be elucidated from single-cell mRNA sequencing of CTCs. Citation Format: Naveen Ramalingam, Yi Fang Lee, Lukasz Szpankowski, Anne Leyrat, Brian Fowler, Jovina Tan, Chong Tracy Lu, Ninez Delos Angeles, Chad Sanada, Cassandra Greene, Kyle Hukari, Andrew Wu, Yoon-Sim Yap, Jay West, Ali Asgar Bhagat. Label-free enrichment and integrated full-length mRNA transcriptome analysis of single live circulating tumor cells from breast cancer patients [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2923. doi:10.1158/1538-7445.AM2017-2923


Leukemia | 2018

A human bone marrow mesodermal-derived cell population with hemogenic potential

Saloomeh Mokhtari; Evan Colletti; Weihong Yin; Chad Sanada; Zanetta S. Lamar; Paul J. Simmons; Steven Walker; Colin E. Bishop; Anthony Atala; Esmail D. Zanjani; Christopher D. Porada; Graca Almeida-Porada

The presence, within the human bone marrow, of cells with both endothelial and hemogenic potential has been controversial. Herein, we identify, within the human fetal bone marrow, prior to establishment of hematopoiesis, a unique APLNR+, Stro-1+ cell population, co-expressing markers of early mesodermal precursors and/or hemogenic endothelium. In adult marrow, cells expressing similar markers are also found, but at very low frequency. These adult-derived cells can be extensively culture expanded in vitro without loss of potential, they preserve a biased hemogenic transcriptional profile, and, upon in vitro induction with Oct4, assume a hematopoietic phenotype. In vivo, these cells, upon transplantation into a fetal microenvironment, contribute to the vasculature, and generate hematopoietic cells that provide multilineage repopulation upon serial transplantation. The identification of this human somatic cell population provides novel insights into human ontogenetic hematovascular potential, which could lead to a better understanding of, and new target therapies for, malignant and nonmalignant hematologic disorders.


Scientific Reports | 2017

Single-cell protein-mRNA correlation analysis enabled by multiplexed dual-analyte co-detection

Haibiao Gong; Xiaohui Wang; Benjamin Liu; Stéphane C. Boutet; Ilona Holcomb; Gajalakshmi Dakshinamoorthy; Aik Ooi; Chad Sanada; Gang Sun; Ramesh Ramakrishnan

We have investigated the correlation between proteins and mRNAs in single cells employing an integrated workflow for dual-analyte co-detection. This is achieved by combining the oligo extension reaction (OER), which converts protein levels to DNA levels, with reverse transcription for mRNA detection. Unsupervised gene expression profiling analysis, including principal component analysis and hierarchical clustering, revealed different aspects of the protein-mRNA relationship. Violin plot analysis showed that some genes exhibited similar distribution patterns for proteins and mRNAs. We also demonstrate that cells can be separated into subpopulations based on their protein-mRNA expression profiles, and that different subpopulations have distinct correlation coefficient values. Our results demonstrated that integrated investigations of mRNA and protein levels in single cells allows comprehensive analysis not attainable at bulk levels.


Cancer Research | 2016

Abstract LB-327: Isolation and mRNA-seq analysis of single CTCs from blood samples using an integrated fluidic circuit for functional single cell studies

Lukasz Szpankowski; Gayatri Premasekharan; Naveen Ramalingam; Chad Sanada; Anne A. Leyrat; Brian Fowler; Matthew Edwards; Cassandra Greene; Ilona Holcomb; Charles J. Ryan; Pamela L. Paris; Jay A. West

As the precursor to metastatic cancer, circulating tumor cells (CTCs) hold tremendous potential for increasing our understanding of tumor metastasis, advancing personalized therapeutics, and have emerged as a significant source of genetic material for clinical guidance. Recent technical advances have enabled the enrichment and genomic characterization of CTCs at the single cell level and have revealed significant and clinically relevant heterogeneity in these rare cells. However, data is lacking for existing methods to link the functional differences of single CTCs to their underlying genomic structure and activity. We have developed an approach which integrates key aspects of biologically based experimentation at the single cell level onto an integrated fluidic circuit. This device allows for the selection and isolation of single CTCs in a size-independent fashion from a pre-enriched population based on fluorescent markers where they can be individually cultured and exposed to a variety of drugs/reagents under environmentally controlled conditions or processed directly for mRNA-seq. If desired, an extracellular matrix can be deposited into each of the single cell culture chambers. Cell activity and phenotype can be monitored in-situ in response to treatment conditions, followed by preparation of the mRNA transcriptome for RNA-seq analysis. As the entire workflow following pre-enrichment is integrated onto a single microfluidic chip, very little manipulation of the single cell is required, serving to preserve the mRNA signature of each cell. To optimize and validate the workflow, PC3 cell lines were spiked-in to blood from healthy donors followed by pre-enrichment for metastatic cancer cells using commercially available CTC isolation platforms. Our microfluidic device further purified and isolated the desired single PC3 cells, and prepared the cells for expression analysis via RNA-seq. The sequencing results further validated the ability of our workflow to isolate rare metastatic cancer cells from human blood and moreover confirmed highly correlated gene expression profiles as compared to cultured PC3 population controls (r = 0.89 across 23,562 genes) and non spike-in single cell samples (r = 0.90 across 23,562 genes). We applied this workflow to the blood of metastatic castration resistant prostate cancer (mCRPC) patients and obtained single cell cDNA from the isolated CTCs from which we were able to generate sequencing libraries. Future work using this system will help establish a robust methodology to enable more complex perturbation and functional studies of single cells from clinical samples. Citation Format: Lukasz Szpankowski, Gayatri Premasekharan, Naveen Ramalingam, Chad Sanada, Anne Leyrat, Brian Fowler, Matthew Edwards, Cassandra Greene, Ilona Holcomb, Charles J. Ryan, Pamela L. Paris, Jay A.A. West. Isolation and mRNA-seq analysis of single CTCs from blood samples using an integrated fluidic circuit for functional single cell studies. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr LB-327.


Cancer Research | 2017

Abstract 1847: Evaluating the metastatic potential and the molecular heterogeneity of patient-derived orthotopic xenograft models of triple-negative breast cancer

Vishnu C. Ramani; Rakhi Gupta; Gerald Quon; Melanie Triboulet; Corinne Renier; Cassandra Greene; Chad Sanada; Tracy Lu; Lukasz Szpankowski; Naveen Ramalingam; Ameen A. Salahudeen; Sean de la O; Ranjani Rajapaksa; Shoshana Levy; Anne A. Leyrat; Jay A. West; Elodie Sollier-Christen; Calvin J. Kuo; George W. Sledge; Stefanie S. Jeffrey


Blood | 2010

Phenotypic Correction of Hemophilia A by Postnatal Intraperitoneal Transplantation of FVIII-Expressing MSC

Christopher D. Porada; Chad Sanada; Eason Kuo; Evan Colletti; Robert Moot; Christopher B. Doering; H. Trent Spencer; Graça Almeida-Porada

Collaboration


Dive into the Chad Sanada's collaboration.

Top Co-Authors

Avatar

Christopher D. Porada

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graça Almeida-Porada

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar

Naveen Ramalingam

Nanyang Technological University

View shared research outputs
Top Co-Authors

Avatar

Paul J. Simmons

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar

Saloomeh Mokhtari

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Graca Almeida-Porada

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge