DRB Faculty

Regular Graduate Faculty

Vernadeth B. Alarcon, PhDAlarcon, Vernadeth B.

Affiliation: School of Medicine, Institute for Biogenesis Research
Position: Associate Professor
Degree: PhD (University of Toronto, Canada)
Phone: (808) 692 1417
Fax: (808) 692 1962
Email: vernadet@hawaii.edu 
Address: 651 Ilalo St., Biosciences Building, Room 163J/H, Honolulu, HI 96813


Research projects:

Preimplantation embryo, Cell lineage, Cell polarity, Developmental toxicants

Description of research:

Mammals, such as ourselves, are unique among other organisms in that our embryonic phase needs to implant into the mother's uterus and create the placenta to allow further development. To achieve such unique style of development, we have evolved to generate a special type of cells, called the trophectoderm, during the very early stages of embryo development (Figures 1 and 2). Trophectoderm is dedicated for implantation and placenta formation. In fact, the first and critical decision that embryonic cells must make after fertilization is whether they commit to become part of the future body or the trophectoderm, i.e., the precursor of placenta. This first decision needs to be controlled very carefully in a balanced manner because embryos cannot survive or develop further if one cell type (either "future body cells", called the inner cell mass, or "future placenta cells", the trophectoderm) is not generated sufficiently.

Our lab's research goal is to uncover the molecular mechanisms that control this early cell type decision-making, using mouse embryos as well as mouse and human cell lines. We have identified candidate genes, including cell polarity regulators (e.g., Alarcon 2010; Kono et al. 2014), and we are testing their roles in the embryo. We employ various experimental approaches, such as in vitro embryo culture, microinjection of the embryo, chimera production, embryo transfer into surrogate mother's reproductive tract, and cell and molecular biology techniques. Understanding the mechanisms of cell type decision-making in the early embryo has applications in the treatment of infertility, especially in ART (assisted reproductive technologies). Our findings have potential to serve as a basis for improving in vitro culture conditions of human embryos and for identifying healthy embryos for uterine transfer to produce successful pregnancies.

embryo1.png

Figure 1: A preimplantation mouse embryo developing over time (left to right): 2-cell, 8-cell, early blastocyst, and late blastocyst. In the late blastocyst, cells have committed to the trophectoderm (future placenta) and inner cell mass (future body) lineages. Live development in vitro was recorded for three days, using time-lapse video microscopy.

blastocysts.png

Figure 2: Immunofluorescently stained blastocysts, showing expression of transcription factors essential for cell-type commitment. (A) OCT4 (green) protein is localized in inner cell mass. (B) CDX2 (yellow) protein is localized in trophectoderm. Nuclei are stained red with propidium iodide. Images were taken using confocal microscopy.


Selected publications:

  • Alarcon, V.B., Marikawa, Y. (2016) Statins inhibit blastocyst formation by preventing geranylgeranylation. Molecular Human Reproduction [Epub ahead of print]
  • Kono, K., Tamashiro, D.A., Alarcon, V.B. (2014) Inhibition of RHO-ROCK signaling enhances ICM and suppresses TE characteristics through activation of Hippo signaling in the mouse blastocyst. Developmental Biology 394, 142-155.
  • Laeno, A.M., Tamashiro, D.A., Alarcon, V.B. (2013) Rho-associated kinase activity is required for proper morphogenesis of the inner cell mass in the mouse blastocyst. Biology of Reproduction 89, 122.
  • Hirate, Y., Hirahara, S., Inoue, K., Suzuki, A., Alarcon, V.B., Akimoto, K., Hirai, T., Hara, T., Adachi, M., Chida, K., Ohno, S., Marikawa, Y., Nakao, K., Shimono, A., Sasaki, H. (2013) Polarity-dependent distribution of angiomotin localizes Hippo signaling in preimplantation embryos. Current Biology 23, 1181-1194.
  • Marikawa, Y., Alarcon, V.B. (2012) Creation of trophectoderm, the first epithelium, in mouse preimplantation development. Results and Problems in Cell Differentiation 55, 165-184. Book chapter.
  • Alarcon, V.B. (2010) Cell polarity regulator PARD6B is essential for trophectoderm formation in the preimplantation mouse embryo. Biology of Reproduction 83, 347-358.

Commonly requested Alarcon's lab protocols:

  • Immunofluorescent staining of preimplantation embryos (under construction)

Other publications:

  • Marikawa, Y., Alarcon, V.B. (2009) Establishment of trophectoderm and inner cell mass lineages in the mouse embryo. Molecular Reproduction and Development 76, 1019-1032.
  • Marikawa, Y., Tamashiro, D.A., Fujita, T.C., Alarcon, V.B. (2009) Aggregated P19 mouse embryonal carcinoma cells as a simple in vitro model to study the molecular regulations of mesoderm formation and axial elongation morphogenesis. Genesis 47, 93-106.
  • Alarcon, V.B., Marikawa, Y. (2008) Spatial alignment of the mouse blastocyst axis across the first cleavage plane is caused by mechanical constraint rather than developmental bias among blastomeres. Molecular Reproduction and Development 75, 1143-1153.
  • Tamashiro, D.A., Alarcon, V.B., Marikawa, Y. (2008) Ectopic expression of mouse Sry interferes with Wnt/beta-catenin signaling in mouse embryonal carcinoma cell lines. Biochimica Biophysica Acta 1780, 1395-1402.
  • Fogelgren, B., Kuroyama, M.C., McBratney-Owen, B., Spence, A.A., Malahn, L.E., Anawati, M.K., Cabatbat, C., Alarcon, V.B., Marikawa, Y., Lozanoff, S. (2008) Misexpression of Six2 is associated with heritable frontonasal dysplasia and renal hypoplasia in 3H1 Br mice. Developmental Dynamics 237, 1767-1779.
  • Yamazaki, Y., Fujita, T.C., Low, E.W., Alarcon, V.B., Yanagimachi, R., Marikawa, Y. (2006) Gradual DNA demethylation of the Oct4 promoter in cloned mouse embryos. Molecular Reproduction and Development 73, 180-188.
  • Hiiragi, T., Alarcon, V.B., Fujimori, T., Louvet-Vallee, S., Maleszewski, M., Marikawa, Y., Maro, B., Solter, D. (2006) Where do we stand now? Mouse early embryo patterning meeting in Freiburg, Germany (2005). International Journal of Developmental Biology 50, 581-588.
  • Alarcon, V.B., Marikawa, Y. (2005) Unbiased contribution of the first two blastomeres to mouse blastocyst development. Molecular Reproduction and Development 72, 354-361.
  • Marikawa, Y., Fujita, T.C., Alarcon, V.B. (2005) Heterogeneous DNA methylation status of the regulatory element of the mouse Oct4 gene in adult somatic cell population. Cloning and Stem Cells 7, 8-16.
  • Alarcon, V.B., Marikawa, Y. (2004) Molecular study of mouse peri-implantation development using the in vitro culture of aggregated inner cell mass. Molecular Reproduction and Development 67, 83-90.
  • Marikawa, Y., Fujita, T.C., Alarcon, V.B. (2004) An enhancer-trap LacZ transgene reveals a distinct expression pattern of Kinesin family 26B in mouse embryos. Development Genes and Evolution 214, 64-71.
  • Alarcon, V.B., Marikawa, Y. (2003) Deviation of the blastocyst axis from the first cleavage plane does not affect the quality of mouse postimplantation development. Biology of Reproduction 69, 1208-1212.
  • Alarcon, V.B., Elinson, R.P. (2001) RNA anchoring in the vegetal cortex of the Xenopus oocyte. Journal of Cell Science 114, 1731-1741.
  • Alarcon, V.B., Filosa, M.F., Youson, J.H. (1997) Cytokeratins in the liver of the sea lamprey (Petromyzon marinus) before and after metamorphosis. Cell and Tissue Research 287, 365-374.
  • Hudson, J.W., Alarcon, V.B., Elinson, R.P. (1996) Identification of new localized RNAs in the Xenopus oocyte by differential display PCR. Developmental Genetics 19, 190-198.
  • Alarcon, V.B., Filosa, M.F., Youson, J.H. (1994) Keratin polypeptides in the epidermis of the larval (ammocoete) sea lamprey, Petromyzon marinus L., show a cell type-specific immunolocalization. Canadian Journal of Zoology 72, 190-194.

Richard Allsopp, PhDRichard Allsopp

Affiliation: School of Medicine, Institute for Biogenesis Research
Position: Assistant Professor
Degree: PhD
Phone: (808) 692 1412
Fax: (808) 692 1951
Email: allsopp@hawaii.edu 
Address: 651 Ilalo St., BSB 163B, Honolulu, HI 96813


Research projects:

Analysis of the role of telomerase in stem cells and human diseases.

Description of research:

Telomerase is required to maintain telomeres, the protective cap at the end of chromosomes, in all eukaryotic cells. In mammals, including humans, some somatic cells in adults lack telomerase, and as a consequence, telomeres gradually short during cell division and as a function of age. Stem cells are required to replenish dead or damaged cells throughout life, and therefore telomerase is thought to play an important role in stem cell survival. While telomerase is indeed detectable in many types of human stem cells (unlike mature somatic cells), in some cases, such as in the blood, there isn't sufficient telomerase to maintain telomere length as these cells divide, and as a consequence, telomeres shorten in all blood cells during aging. On the other hand, male germ cells do have sufficient telomerase to thwart telomere loss during aging. one of the primary goals in my lab is to get a better understanding as to how telomerase activity levels are regulated in different types of stem cells. Recently, we have performed a screen for transcriptional regulators in embryonic stem cells and found that the transcription factor Hypoxia Inducible Factor 1 alpha (Hif1alpha) is essential for maintainence of functional levels of telomerase in these cells. One of the future goals of our work is get a better understanding as to the role of Hif1alpha in regulating telomerase in other types of stem cells. My lab is also interested in evaluating the therapeutic potential of using stem cells, particularly stem cells derived from the bone marrow, which is a pracxtical source of stem cells in adults, to treat cardiac infarcts using a murine model system.


Selected publications:

  • Allsopp, R. C., Vaziri, H., Patterson, C., Goldstein, S., Younglai, E. V., Futcher, A. B., Greider, C. W. and Harley, C. B. Telomere length predicts replicative capacity of human fibroblasts. 1992. Proc. Nat. Acad. Sci. U. S. A. 89, 10114-10118.
  • Harley, C. B., Vaziri, H., Counter, C. and Allsopp, R. Telomere hypothesis of cell aging. 1992. Exp. Geront. 27, 375-382.
  • Vaziri, H., Dragowska, W., Allsopp, R. C., Thomas, T. E., Harley, C. B. and Lansdorp, P. Evidence for a mitotic clock in primative blood stem cells: Loss of telomeric DNA with age. 1994. Proc. Nat. Acad. Sci. U.S.A. 91, 9857-9860.
  • Allsopp, R. C. and Harley, C. B. Evidence for a critical telomere length in senescent human fibroblasts. 1995. Exp. Cell Res. 219, 130-136.
  • Allsopp, R. C., Chang, E. C., Kashefi-Aazam, M., Rogoev, E. and Harley, C. B. Telomere shortening is associated with cell division in vitro and in vivo. 1995. Exp. Cell Res. 220, 194-200.
  • Greenberg, R., Allsopp, R., Chin, L., Morin, G. and dePinho, R. Expression of mouse telomerase reverse transcriptase during development, differentiation and proliferation. 1998. Oncogene, 16, 1723-30.
  • Allsopp, R., Cheshier, S. and Weissman, I.L. Telomere shortening accompanies increased cell cycle activity during serial transplantation of hematopoietic stem cells. 2001. J. Exp. Med. 193, 917-924.
  • Wagers, A.J., Allsopp, R.C. and Weissman, I.L. Changes in integrin expression are associated with altered homing properties of Lin(-/lo)Thy1.1(lo)Sca-1(+)c-kit(+) hematopoietic stem cells following mobilization by cyclophosphamide/granulocyte colony-stimulating factor. 2002. Exp Hematol. 30, 176-185.
  • Allsopp, R. and Weissman, I.L. Replicative senescence of hematopoietic stem cells during serial transplantation: does telomere shortening play a role? 2002. Oncogene. 13, 3270-3273.
  • Allsopp, R., Cheshier, S. and Weissman, I.L. Telomerase activation and rejuvenation of telomere length in activated of T cells derived from hematopoietic stem cells following serial transplantation. 2002. J. Exp. Med. 196, 1427-33.
  • Allsopp, R., Morin, G., DePinho, R., Harley, C. and Weissman, I.L. Telomerase is required to slow telomere shortening and extend replicative lifespan of HSC during serial transplantation. 2003. Blood. 102, 517-20.
  • Allsopp, R., Morin, G., DePinho, R., Harley, C. and Weissman, I.L. Effect of TERT over-expression on the long-term transplantation capacity of hematopoietic stem cells. 2003. Nat. Med. 9, 369-71.
  • Coussens, M., Yamazaki, Y., Moisyadi, S., Suganuma, R., Yanigimachi, R. and Allsopp, R. Regulation and effects of modulation of telomerase reverse transcriptase expression in primordial germ cells during development.2006. Biol. Reprod. 75, 785-791.
  • Allsopp, R., Shimoda, J., Easa, D., and Ward, K. Long telomeres in the mature human placenta. 2007. Placenta. 28, 324-7.
  • Narala S., Allsopp, R. et al. SIRT1 acts as a nutrient-sensitive growth suppressor and its loss is associated with increased AMPK and telomerase activity. 2008. Mol Cell Biol. 19, 1210-19.
  • Coussens M, Maresh G, Yanagimachi R, Maeda G, and Allsopp R. Sirt1 Deficiency attenuates spermatogenesis and germ cell function. 2008. PLoS ONE. 13, e1571-74.
  • Davy, P and Allsopp R. Balancing out the ends during iPSC nuclear reprogramming. 2009. Cell Stem Cell. 4, 95-96.
  • Davy P, Nagata M, Bullard P, Fogelson NS, Allsopp R. 2009. Fetal growth restriction is associated with accelerated telomere shortening and increased expression of cell senescence markers in the placenta. Placenta, In Press.
  • Squires J, Davy P, Berry M, Allsopp R. 2009. Attenuated expression of SECIS binding protein 2 causes loss of telomeric reserve without affecting telomerase. Exp Geront. In Press.
  • Meznikova M, Allsopp R, Erdmann N, Weissman I, Harrington LH. 2009. Telomere re-equilibration after prolonged telomerase heterozygosity in mice. Disease Models and Mechanisms. In Press.

Ben Fogelgren, PhDBen Fogelgren

Affiliation: School of Medicine, Department of Anatomy, Biochemistry, and Physiology
Position: Assistant Professor
Degree: PhD (University of Hawaiʻi)
Phone: (808) 692-1420
Fax: (808) 692-1951
Email: fogelgre@hawaii.edu 
Address: 651 Ilalo St., BSB 110, Honolulu, HI 96813


Research projects:

Molecular mechanisms of primary cilia assembly and signaling.
Genetic regulation of kidney development, physiology, and disease.

Description of research:

Our research is focused on the molecular and genetic causes of abnormal kidney development, as well as the novel causes and treatments of adult renal diseases such as polycystic kidney disease and acute renal injury. The same molecular signals and cellular morphogenesis patterns that we study in the kidney can also give us novel insights into development of other tissues, as well as teach us how these processes can be disrupted in a large variety of diseases.

Cysts and tubules are primary building blocks of the kidney, and defects in cystogenesis or tubulogenesis during kidney development can result in a spectrum of pediatric and adult renal diseases. Relevant to our research, during kidney development, disruption in the formation of de novo epithelial cysts from pools of mesenchymal stem cells lead to various forms of congenital abnormalities such as kidney hypoplasia, dysplasia, or agenesis. On the other hand, following nephrogenesis, when the inhibitory signals that halt excessive cyst formation are disturbed, forms of renal disease such as polycystic kidney disease (PKD) can occur. Autosomal dominant PKD (ADPKD), which affects approximately 500,000 Americans and currently has no approved treatment, is the most common potentially lethal genetic disease. Every variation of human PKD has been attributed to mutations in genes important to the assembly or function of the primary cilium, a thin rod-like organelle on renal tubular epithelial cells which projects into the tubular lumen (See Figure 1).

Primary cilia have been found on the surface of most growth-arrested or differentiated mammalians cells, including a large variety of epithelial cells, endothelial cells, connective-tissue and muscle cells, as well as neurons and even embryonic stem cells. Although the presence of primary cilia has been noted for over a hundred years, its biological function was a mystery. Due to an exploding volume of research in just the last decade, it is now believed the primary cilium acts as a "sensory antenna" to relay signals to the cell based on the extracellular environment. These can include sensory clues such as mechanical stimulation (bending or rotation), chemosensation (detection of growth factors, morphogens, etc.), osmolarity, light, temperature, and even gravitational pull. It is thought that primary cilia along the renal tubules are responsible for detecting fluid composition and flow dynamics, and when defective, the cells misinterpret this as a signal to dedifferentiate and proliferate, resulting in the formation of large pathogenic renal cysts. However, defects in primary cilia can affect other tissues as well, and have resulted in a new classification of human disorders termed "ciliopathies".

Currently, we have research projects focused both on the molecular mechanisms of de novo cyst formation, and on primary cilia assembly and signaling. We have found both of these cellular activities depend on the exocyst complex, a highly conserved eight-protein complex (see Figure 2), which is involved in targeted secretory vesicle transport. We are working hard to discover how cells regulate the exocyst subunits in order to direct polarized trafficking to specific locales such as primary cilia and the lumens of growing epithelial cysts. These discoveries may lay the groundwork for novel therapies for adult and pediatric renal diseases, and expand our knowledge of some of the basic cellular and molecular mechanisms by which the human body controls its development.

primary cilium
Figure 1: The primary cilium, found on almost all cell types, is a single rod-like organelle that extends out into the extracellular environment to relay information back to the cell. A. In polarized epithelial cells, the primary cilium extends out from the apical surface into the luminal space. All primary cilia are non-motile, comprised of 9 microtubule doublets lacking a central pair, extending from a basal body. B. Scanning electronic micrograph of the lumen-facing side of renal tubular epithelial cells displaying long primary cilia (from Kessel and Kardon, Tissues And Organs, 1979). C. Immunostaining of exocyst subunit Sec8 (shown in red) and polycystin-2 (the protein encoded by Pkd2 gene, shown in green) along primary cilia in cultured renal tubular epithelial cells.

 

plasma membrane
Figure 2: The exocyst is a highly-conserved complex that guides intracellular vesicles to specific sites of exocytosis (figure adapted from Lipschutz and Mostov, 2002), and is composed of eight proteins (Sec3, Sec5, Sec6, Sec8, Sec10, Sec15, Exo70, and Sec84). Regulation of the exocyst subunits is a method by which the cell can control both the timing and subcellular location of integral protein (and membrane) delivery to the cell's plasma membrane. For example, a subunit of the exocyst is phosphorylated in response to insulin signaling in adipocytes, which induces exocytosis of vesicles carrying the Glut4 transporter to allow intake of glucose (Inoue et al., Nature, 2002).

 


Selected publications:

  • Polgar N, Lee AJ, Lui VH, Napoli JA, Fogelgren B. The exocyst gene Sec10 regulates renal epithelial monolayer homeostasis and apoptotic sensitivity. Am J Physiol Cell Physiol. 2015 Aug 1;309(3):C190-201.
  • Fogelgren B, Polgar N, Lui VH, Lee AJ, Tamashiro KK, Napoli JA, Walton CB, Zuo X, Lipschutz JH. Urothelial Defects from Targeted Inactivation of Exocyst Sec10 in Mice Cause Ureteropelvic Junction Obstructions. PLoS One. 2015 Jun 5;10(6):e0129346.
  • Fogelgren B, Zuo X, Buonato JM, Vasilyev A, Baek JI, Choi SY, Chacon-Heszele MF, Palmyre A, Polgar N, Drummond I, Park KM, Lazzara MJ, Lipschutz JH. Exocyst Sec10 protects renal tubule cells from injury by EGFR/MAPK activation and effects on endocytosis. Am J Physiol Renal Physiol. 2014 Dec 15;307(12):F1334-41.
  • Choi SY, Fogelgren B, Zuo X, Huang L, McKenna S, Lingappa VR, Lipschutz JH. Exocyst Sec10 is involved in basolateral protein translation and translocation in the endoplasmic reticulum. Nephron Exp Nephrol. 2012;120(4):e134-40.
  • Fogelgren B, Lin SY, Zuo X, Jaffe KM, Park KM, Reichert RJ, Bell PD, Burdine RD, and Lipschutz JH. Exocyst Sec10 interacts with polycystin-2 and knockdown causes PKD-phenotypes. PLoS Genetics, 7(4): e1001361, doi:10.1371/journal.pgen.1001361, 2011.
  • Chung DC, Fogelgren B, Park KM, Heidenberg J, Zuo XF, Huang L, Bennett J, and Lipschutz JH. Adeno-associated virus (AAV)-mediated gene transfer to renal tubule cells via a retrograde ureteral approach. Nephron EXTRA, 1: 217-223, 2011.
  • Somponpun J, Wong B, Kuroyama M, Hynd T, Fogelgren B, and Lozanoff S. Osmoregulatory defect associated with hypodysplastic kidney in 3H1 Brachyrrhine mice. American Journal of Physiology: Regulatory, Integrative and Comparative Physiology, 301(3): R682-R689, 2011.
  • Zuo XF, Fogelgren B, and Lipschutz JH. The small GTPase cdc42 is necessary for primary ciliogenesis in renal tubular epithelial cells. Journal of Biological Chemistry, 286(25): 22469-22477, 2011.
  • Park KM, Fogelgren B, Zuo X, Kim J, Chung DC, Lipschutz JH. Exocyst Sec10 protects epithelial barrier integrity and enhances recovery following oxidative stress, by activation of the MAPK pathway. American Journal of Physiology: Renal Physiology, 298(3): F818-F826, 2010.
  • Fogelgren B, Yang S, Sharp IC, Huckstep OJ, Ma W, Somponpun J, Carlson EC, Uyehara C and Lozanoff S. Deficiency in Six2 during prenatal development is associated with reduced nephron number, chronic renal failure, and hypertension in Br/+ adult mice. American Journal of Physiology: Renal Physiology, 296(5): F1166-F1178, 2009.
  • Fogelgren B, Kuroyama MC, McBratney-Owen B, Spence AA, Malahn LE, Anawati MK, Cabatbat C, Alarcon VB, Marikawa Y and Lozanoff S. Misexpression of Six2 is associated with heritable frontonasal dysplasia and renal hypoplasia in 3H1 Br mice. Developmental Dynamics, 237(7):1767-79, 2008.
  • Polgar N, Fogelgren B, Shipley JM and Csiszar K. Lysyl oxidase interacts with hormone placental lactogen and synergistically promotes breast epithelial cell proliferation and migration. Journal of Biological Chemistry, 170(2): 578-589, 2007.
  • Cao T, Racz P, Szauter KM, Groma G, Fogelgren B, Pankotai E, He QP and Csiszar K. Mutation in Mpzl3, a novel gene encoding a predicted adhesion protein, in the Rough Coat (rc) mice with severe skin and hair abnormalities. Journal of Investigative Dermatology, 127(6): 1375-86, 2007.
  • Payne SL, Fogelgren B, Hess AR, Seftor EA, Wiley EL, Fong SF, Csiszar K, Hendrix MJ and Kirschmann DA. Lysyl oxidase regulates breast cancer cell migration and adhesion through a hydrogen peroxide-mediated mechanism. Cancer Research, 65(24): 11429-36, 2005.
  • Fogelgren B, Polgar N, Szauter KM, Ujfaludi Z, Laczko R, Fong KS and Csiszar K. Cellular fibronectin binds to lysyl oxidase with high affinity and is critical for its proteolytic activation. Journal of Biological Chemistry, 280(26): 24690-97, 2005.

Jason Higa

Affiliation: School of Medicine, Department of Anatomy, Biochemistry, and Physiology
Position: Assistant Professor
Phone: (808) 692-1009
Fax: (808) 692-5497
Email: jkenjih@hawaii.edu 
Address: 651 Ilalo St., MEB 307R, Honolulu, HI 96813

Scott Lozanoff

Affiliation: School of Medicine, Department of Anatomy, Biochemistry, and Physiology
Position: Professor
Phone: (808) 692-1442
Fax: (808) 692-1951
Email: lozanoff@hawaii.edu 
Address: 651 Ilalo St., BSB 119A, Honolulu, HI 96813

 

Yusuke Marikawa, PhDYusuke Marikawa, PhD

Affiliation: School of Medicine, Institute for Biogenesis Research
Position: Associate Professor
Degree: PhD (Kyoto University, Japan)
Phone: (808) 692-1411
Fax: (808) 692-1962
Email: marikawa@hawaii.edu 
Address: 651 Ilalo St., BSB 163A, Honolulu, HI 96813


Research projects:

Axis Specification and Germ Layer Formation in Mammalian Embryos.
Differentiation and Morphogenesis in Pluripotent Stem Cells.
Stem Cell-Based Detection of Teratogens.

Description of research:

How can a single cell, like the fertilized egg, transform into a complex architecture, like our body? With respect to this biggest question of Developmental Biology, my lab is currently conducting research projects under the following two main themes:

"Axis Specification and Germ Layer Formation in Mammalian Embryos"

Soon after implantation, the embryo of human or mouse is a single layer of epithelial tissue, called the epiblast. Later, one edge of the epiblast forms the structure called the primitive streak, from which cells migrate away to form new tissue layers. This event, i.e., the formation of the primary germ layers (ectoderm, mesoderm, and endoderm), is the first step towards the generation of complex body patterns. Furthermore, the primitive streak formation is the first morphological landmark of body axis specification, as it establishes the caudal end of the future body.

Activation of Wnt/beta-catenin signaling, an evolutionary conserved signal transduction machinery, is the key to the formation of the primitive streak in the epiblast. The specific questions that are addressed in my current projects are:

1) How Wnt/beta-catenin signaling is activated in a localized region of the epiblast?
2) What genes are turned on by Wnt/beta-catenin signaling in the primitive streak?
3) How can cells in the primitive streak transform from epithelial into migratory state?

"Differentiation and Morphogenesis in Pluripotent Stem Cells"

Embryonic stem (ES) cells and induced pluripotent stem (iPS) cells can be maintained and propagated in a culture dish as undifferentiated cell populations, which are very similar to early embryonic cells before germ layer formation. Because studies of mammalian embryos are often hindered by the unique reproductive mode of mammals (i.e., embryos normally develop in the uterus), pluripotent stem cells, like ES and iPS cells, can serve as great in vitro models, which can be experimentally manipulated more easily. Furthermore, these pluripotent stem cells offer tremendous promise for the future regenerative medicine, as they can be used to derive various types of functional tissues in vitro, and those stem cell-derived tissues may be used for tissue replacement therapy and drug screening.

The goal of my lab is to elucidate molecular and cellular mechanisms of differentiation (e.g., of mesoderm and endoderm tissues) and morphogenesis (e.g., of tissue elongation along the body axis) using mouse and human pluripotent stem cells.

confocal microscopy
Figure1: Confocal microscopy image of mouse E6.5 stage embryo, showing the distribution of pluripotency regulator OCT4 protein (green) and all the nuclei (red). The epiblast shows nuclear localization of OCT4 protein. Arrowhead indicates the site of the primitive streak formation.

 

in vitro axial
Figure 2: in vitro axial elongation morphogenesis in aggregated P19 embryonal carcinoma cells. Scale bar = 1 mm.

 


Selected publications:

  • Yuan CJ, Marikawa Y. (2017) Developmental toxicity assessment of common excipients using a stem cell-based in vitro morphogenesis model. Food and Chemical Toxicology. 109:376-385.
  • Warkus EL, Marikawa Y. (2017) Exposure-based validation of an in vitro gastrulation model for developmental toxicity sssays. Toxicological Sciences. 157:235-245.
  • Li AS, Marikawa Y. (2016) Adverse effect of valproic acid on an in vitro gastrulation model entails activation of retinoic acid signaling. Reproductive Toxicology. 66:68-83.
  • Warkus EL, Yuen AA, Lau CG, Marikawa Y.Use of in vitro morphogenesis of mouse embryoid bodies to assess developmental toxicity of therapeutic drugs contraindicated in pregnancy. Toxicological Sciences. 149:15-30.
  • Li AS, Marikawa Y. (2015) An in vitro gastrulation model recapitulates the morphogenetic impact of pharmacological inhibitors of developmental signaling pathways. Molecular Reproduction and Development. 82:1015-1036.
  • Lau CG, Marikawa Y. (2014) Morphology-based mammalian stem cell tests reveal potential developmental toxicity of donepezil. Molecular Reproduction and Development. 81:994-1008.
  • Jaremko KL, Marikawa Y. (2013) Regulation of Developmental Competence and Commitment towards the Definitive Endoderm Lineage in Human Embryonic Stem Cells. Stem Cell Research. 10:489-502.
  • Hirate Y, Hirahara S, Inoue K, Suzuki A, Alarcon VB, Akimoto K, Hirai T, Hara T, Adachi M, Chida K, Ohno S, Marikawa Y, Nakao K, Shimono A, Sasaki H. (2013) Polarity-dependent distribution of angiomotin localizes Hippo signaling in preimplantation embryos. Current Biology 23:1181-1194.
  • Tamura AN, Huang TT, Marikawa Y. (2013) Impact of vitrification on the meiotic spindle and components of the microtubule-organizing center in mouse mature oocytes. Biology of Reproduction 89:112
  • Tamashiro DA, Alarcon VB, Marikawa Y. (2012) Nkx1-2 is a transcriptional repressor and is essential for the activation of Brachyury in P19 mouse embryonal carcinoma cell. Differentiation. 83:282-292.
  • Marikawa Y, Alarcon VB. (2012) Creation of trophectoderm, the first epithelium, in mouse preimplantation development. Results and Problems of Cell Differentiation 55:165-184.
  • Complete List of Published Work in MyBibliography.

Selected publications:

Takashi MatsuiTakashi Matsui

Affiliation: School of Medicine, Dept. of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research
Position: Professor and Chair
Degree: MD and PhD (Jikei Univ Sch of Med, JAPAN)
Phone: (808) 692-1554
Fax: (808) 692-1973
Email: tmatsui@hawaii.edu  
Address: 651 Ilalo St., BSB 110, Honolulu, HI 96813


Research projects:

The role of mammalian target of rapamycin (mTOR) in the heart, Cardiac cell signaling controlling cell survival, Diabetic hearts, Ferroptosis

Description of research:

Our research is focused on the insulin signaling pathway in cardiomyocytes, especially cardioprotective effects against pathological settings such as ischemia. Our interests center around the role of the mechanistic target of rapamycin (mTOR), which is intimately related to the insulin/phosphatidylinositol 3-kinase (PI3K)/Akt signal transduction pathway. In order to investigate the role of mTOR in the heart, we utilize a variety of in vitro, in vivo, and ex vivo models of heart failure with genetically manipulated models of mTOR such as transgenic and knockout mice. We have reported that the role of cardiac mTOR in prevention of cardiomyocyte cell death that arises from myocardial infarction and cardiac hypertrophy, apparent risk factors for heart failure.

Diabetes is an independent risk factor for both heart failure and ischemic heart disease. Because of the important role of mTOR in insulin signaling, we have been working to determine the role of mTOR in diabetic hearts, and exploring the mTOR signaling pathway as a novel therapeutic target for treatment of heart failure in diabetes.

We recently demonstrated that mTOR is necessary and sufficient for cardiomyocyte protection against iron-mediated cell death that includes excessive iron-induced cell death and ferroptosis that is an iron-dependent form of regulated cell death. This is the first report that ferroptosis is a significant type of cell death in cardiomyocytes. We are currently defining the pathophysiological role of ferroptosis in cardiac diseases such as acute myocardial infarction and heart failure.

ex vivo isolated heart perfusion
Figure1: A. ex vivo isolated heart perfusion (Langendorff perfused heart) B. Isolated adult cardiomyocytes expressed with green fluorescent protein (GFP)

 

intramyocardial fibrosis
Figure 2: Intramyocardial Fibrosis (blue) observed in post-Myocardial Infarction. (Masson's trichome staining)

Key publications: (*Corresponding author)

Peer-reviewed articles:
  • 1. Mishra PK, Adameova A, Hill AJ, Baines CP, Kang P, Dawney JM, Narula J, Takahashi M, Abbate A, Piristine H, Su S, Higa JK, Kawasaki NK, Matsui T. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 317: H891-H922, 2019. doi: 10.1152/ajpheart.00259.2019. PMID: 3141859
  • 2. Yorichika N, Baba Y, Shimada BK, Thakore M, Wong SM, Kobayashi M, Higa JK, Matsui T*. The effects of Tel2 on cardiomyocyte survival. Life Sci. 2019 Sep 1;232:116665. doi: 10.1016/j.lfs.2019.116665. Epub 2019 Jul 16. PMID: 31323273
  • 3. Kobayashi M, Suhara T, Baba Y, Kawasaki KK, Higa JK, Matsui T*. Pathological roles of iron in cardiovascular disease. Curr Drug Targets. 2018;19(9):1068-1076. doi: 10.2174/1389450119666180605112235.
  • 4. Baba Y, Higa JK, Shimada BK, Horiuchi KM, Suhara T, Kobayashi M, Woo JD, Aoyagi H, Marh KS, Kitaoka H, Matsui T*. Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2018;314:H659-H668 [selected in Editor Picked Articles and posted on Podcast]
  • 5. Suhara T, Baba Y, Shimada BK, Higa JK, Matsui T*. The mTOR signaling pathway in myocardial dysfunction in type 2 diabetes mellitus. Curr Diab Rep. 2017;17(6):38.
  • 6. Aoyagi T, Higa JK, Aoyagi H, Yorichika N, Shimada BK, Matsui T*. Cardiac mTOR rescues the detrimental effects of diet-induced obesity in the heart after ischemia-reperfusion. Am J Physiol Heart Circ Physiol. 2015;308(12):H1530-1539.
  • 7. Katz MY, Kusakari Y, Aoyagi H, Higa JK, Xiao C-Y, Abdelkarim AZ, Marh K, Aoyagi A, Rosenzweig A, Lozanoff S, Matsui T*. Three dimensional myocardial scarring along myofibers after coronary ischemia-reperfusion revealed by computerized images of histological assays. Physiol Rep. 2014;2 (7): e12072 (open-access article), doi: 10.14814/phy2.12072.
  • 8. Aoyagi T, Kusakari Y, Xiao C-Y, Inouye BT, Takahashi M, Scherrer-Crosbie M, Rosenzweig A, Hara K, Matsui T*. Cardiac mTOR protects the heart against ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2012;303(1):H75-85.
  • 9. Aoyagi T, Matsui T*. The cardiomyocyte as a source of cytokines in cardiac injury. Journal of Cell Science & Therapy 2011;S5:003 (open-access article). doi:10.4172/2157-7013.S5-003 PMC3594870.
  • 10. Song X, Kusakari Y, Xiao C-Y, Kinsella SD, Rosenberg MA, Scherrer-Crosbie M, Hara K, Rosenzweig A, Matsui T*. mTOR attenuates the inflammatory response in cardiomyocytes and prevents cardiac dysfunction in pathological hypertrophy. Am J Physiol Cell Physiol. 2010;299(6):C1256-66.
  • 11. Matsui T*, Nagoshi T, Hong EG, Luptak I, Hartil K, Li L, Gorovits N, Charron MJ, Kim JK, Tian R, Rosenzweig A. The effects of chronic Akt activation on glucose uptake in the heart. Am J Physiol Endocrinol Metab. 2006;290:E789-97.
Books:
  • 1. Higa JK, Kawasaki NK, Matsui T*. Ferroptosis in Cardiovascular Disease. In: Daolin Tang, editor. Ferroptosis in Health and Disease. Springer Nature; 2019.
  • 2. Matsui T*. Role of Caspases in Apoptotic-Driven Indications. In: Tom O'Brien and Steven D. Linton, editors. Design of Caspase Inhibitors as Potential Clinical Agents. Boca Raton, FL: Taylor & Francis Group; 2008, p59-73.

Alika K. MaunakeaAlika K. Maunakea, Ph.D.

Affiliation: School of Medicine, Department of Native Hawaiian Health
Position: Associate Professor
Graduate Faculty: Molecular Biosciences and Bioengineering (MBBE)
Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Neuroscience Specialization Program

Phone: (808) 692-1048
Fax: (808) 692-1255
Email: amaunake@hawaii.edu  
Address: 651 Ilalo St., Room 222K, Honolulu, HI 96813

Stefan Moisyadi, PhDStefan Moisyadi, PhD

Affiliation: School of Medicine, Institute for Biogenesis Research
Position: Associate Professor
Graduate Faculty: Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 956-3118
Fax: (808) 956-7316
Email: moisyadi@hawaii.edu 
Address: IBR, 1960 East-West Rd. Room E108, Honolulu, HI 96822

Noemi Polgar

Affiliation: School of Medicine, Anatomy, Biochemistry & Physiology
Position: Assistant Professor
Phone: (808) 692-1422
Fax: (808) 692-1951
Email: polgar@hawaii.edu 
Address: 651 Ilalo St., BSB 110, Honolulu, HI 96813

Kathryn J. SchunkeKathryn J. Schunke

Affiliation: Department of Anatomy, Biochemistry and Physiology, Center for Cardiovascular Research, Diabetes Research Center, John A. Burns School of Medicine
Position: Assistant Professor
Degree: PhD 
Phone: (808) 692-1565
Email: kschunke@hawaii.edu 
Address: 651 Ilalo St., BSB 211, Honolulu, HI 96813

Johann Urschitz, PhDJohann Urschitz, PhD

Affiliation: Institute for Biogenesis Research Department of Anatomy,
Biochemistry and Physiology, John A. Burns School of Medicine
Graduate Faculty: Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 956-7417
Fax: (808) 956-7316
Email: johann@hawaii.edu  
Address: IBR, 1960 East-West Rd., Room E112, Honolulu, HI 96822

Monika A. Ward, PhDMonika A. Ward, PhD

Affiliation: Institute for Biogenesis Research Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine
Graduate Faculty: Chair, Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 956-0779
Fax: (808) 956-7316
Email: mward@hawaii.edu   
Address: IBR, 1960 East-West Rd., Room E104, Honolulu, HI 96822

W. Steven Ward, PhDW. Steven Ward, PhD

Affiliation: Institute for Biogenesis Research Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine
Graduate Faculty: Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 956-5189
Fax: (808) 956-7316
Email: wward@hawaii.edu  
Address: IBR, 1960 East-West Rd., Room E108, Honolulu, HI 96822

Yukiko Yamazaki, PhDYukiko Yamazaki, PhD

Affiliation: Institute for Biogenesis Research Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine
Graduate Faculty: Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 692-1416
Fax: (808) 692-1962
Email: yyamazak@hawaii.edu  
Address: 651 Ilalo St., BSB 163-3 Honolulu, HI 96813

Yiqiang ZhangYiqiang Zhang

Affiliation: JABSOM, Center for Cardiovascular Research
Phone: (808) 692-1480
Fax: (808) 692-1973
Email: yiqiang.zhang@hawaii.edu   
Address: 651 Ilalo St., BSB-311D, Honolulu, HI 96813

Cooperating Graduate Faculty

Peter R. Hoffmann, PhD, MSPHPeter R. Hoffmann, PhD, MSPH

peterrh@hawaii.edu  |  (808) 692-1510
Full Member, Cancer Biology Program, University of Hawaiʻi Cancer Center
Academic Appointment(s): Professor, Cell and Molecular Biology Department, John A. Burns School of Medicine, University of Hawaiʻi at Mānoa
Degree(s): PhD, Immunology and Microbiology, University of Colorado
MSPH, Public Health, University of Hawaiʻi at Mānoa

View full bio on the UH Cancer Center website

Thomas Huang, PhDThomas Huang, PhD

Associate Professor
Affiliation: JABSOM, Obstetrics, Gynecology and Women's Health
Email: huangt@hawaii.edu 
Phone: (808) 203-6533
Fax: (808) 955-2174
Address: Kapiolani Med Ctr

Olivier Le Saux, Ph.D. Olivier Le Saux, Ph.D.

Professor and Chair, Department of Cell and Molecular Biology (CMB)
Area of Expertise: Skin and cardiovascular diseases, ectopic calcification
Email: lesaux@hawaii.edu 
Phone: (808) 692-1504

Birendra Mishra PhDBirendra Mishra PhD, MS, DVM

Assistant Professor
Department of Human Nutrition, Food & Animal Sciences, College of Tropical Agriculture and Human Resources – University of Hawai‘i at Mānoa
Email: bmishra@hawaii.edu 
Phone: (808) 956-7021
Fax: (808) 956-4024

Jesse B. Owens, PhDJesse B. Owens, PhD

Affiliation: Institute for Biogenesis Research
Department of Anatomy, Biochemistry and Physiology
John A. Burns School of Medicine
Graduate Faculty: Developmental and Reproductive Biology (DRB)
Cell and Molecular Biology (CMB)
Phone: (808) 956-4828
Fax: (808) 956-7316
Email: jbowens@hawaii.edu 
Address: IBR, 1960 East-West Rd, Room E108, Honolulu, HI 96822

Michelle Tallquist, PhDMichelle Tallquist, PhD

Affiliation: JABSOM, Center for Cardiovascular Research
Position: Associate Professor
Degree: PhD (Immunology, Mayo Clinic and Foundation)
Phone: (808) 692 1579
Fax: (808) 692 1973
Email: michelle.tallquist@hawaii.edu 
Address: BSB 311E, 651 Ilalo Street, Honolulu HI, 96813

Jinzeng Yang, PhDJinzeng Yang, PhD

Affiliation: Department of Human Nutrition, Food and Animal Sciences
Position: Associate Professor
Degree: PhD (University of Alberta, Canada)
Phone: (808) 956 6073
Fax: (808) 956 4024
Email: jinzeng@hawaii.edu 
Address: 1955 East West Road, Rm 216, Honolulu, HI 96822


Masato YoshizawaMasato Yoshizawa

yoshizaw@hawaii.edu 
808-956-6682
LSB 222
Lab Website
Google Scholar

Bradley J. Willcox

Affiliation: Geriatric Medicine
Position: Professor
Phone: (808) 523-8461
Fax: (808) 528-1897
Email: willcox@hawaii.edu 
Address: Kuakini Med Ctr HPM-9


Affiliate Graduate Faculty

ortega_orig-1.webpMichael Ortega, PhD

Principal Investigator
Phone: (808) 691-7902
Fax: (808) 691-7939
miortega@queens.org 

View the Ortega Lab at the Queen's Medical Center website

Claire WrightClaire Wright, PhD

Senior Director, Office of Sponsored Programs
Associate Professor, Biology
School of Natural Science and Mathematics
Email: claire.wright@chaminade.edu
Phone: (808) 739-8343