Network


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

Hotspot


Dive into the research topics where Josiah Ochieng is active.

Publication


Featured researches published by Josiah Ochieng.


Glycoconjugate Journal | 2002

Extracellular functions of galectin-3.

Josiah Ochieng; Vyacheslav Furtak; Pavel Lukyanov

Galectin-3 has been suspected of modulating cell to extracellular matrix interactions in a novel fashion ever since it was first described. However, the rapid accumulation of research data in just the last 8 years alone has completely changed our perspective of this multifunctional protein. Its chimeric nature (consists of carbohydrate recognition and collagen like domains) somehow makes it suited to interact with a plethora of interesting extracellular matrix proteins some of which might enable it to cross the plasma membrane despite its lack of appropriate signal peptides. It is now becoming established as a mediator of signal transduction events on the cell surface as well as a mediator of a variety of extra-cellular processes such as kidney development, angiogenesis, neuronal functions, tumor metastasis, autoimmune disorders, endocytosis and possibly exocytosis. Nevertheless, it still retains its unique position as a mediator/modulator of cell to extracellular matrix adhesive interactions. Cells, particularly epithelial cells which lack galectin-3 expression, interact poorly with their extracellular matrices. In some of these processes, it functions as a matricellular protein, displaying both pro- and anti-adhesive properties. Published in 2004.


Biochimica et Biophysica Acta | 1998

Modulation of the biological functions of galectin-3 by matrix metalloproteinases.

Josiah Ochieng; Brenda Green; Shawnda Evans; Oscar James; Paula Warfield

Galectin-3 is an important intracellular and extracellular lectin which is presumed to interact with extracellular matrix proteins and cell surface glycoproteins in normal and pathophysiological conditions. The exact physiological role of the protein is presently not known. We have previously demonstrated that recombinant human galectin-3 is a novel substrate for metalloproteinases, particularly MMP-2 and MMP-9. These enzymes are capable of efficiently cleaving the Ala62-Tyr63 bond of the ca. 30 kDa galectin-3, generating a 22 kDa fragment with intact carbohydrate recognition domain and a ca. 9 kDa polypeptide comprising the amino terminal end of the intact galectin-3. In this study, we analyzed interactions of the 22 kDa fragment of galectin-3 with immobilized laminins. We have also compared the hemagglutination as well as homodimerization potentials of this fragment with that of intact galectin-3. Our data suggest that cleavage of galectin-3 by metalloproteinases; (a) alters the carbohydrate recognition domain of the lectin so that it binds more tightly to the glycoconjugates and, (b) reduces selfassociation of the galectin molecules thereby abrogating the biological properties dependent on such associations or homodimerization.


PLOS ONE | 2011

Detachment of Breast Tumor Cells Induces Rapid Secretion of Exosomes Which Subsequently Mediate Cellular Adhesion and Spreading

Rainelli Koumangoye; Amos M. Sakwe; J. Shawn Goodwin; Tina Patel; Josiah Ochieng

Exosomes are nano-vesicles secreted by a wide range of mammalian cell types. These vesicles are abundant in serum and other extracellular fluids and contain a large repertoire of proteins, mRNA and microRNA. Exosomes have been implicated in cell to cell communication, the transfer of infectious agents, and neurodegenerative diseases as well as tumor progression. However, the precise mechanisms by which they are internalized and/or secreted remain poorly understood. In order to follow their release and uptake in breast tumor cells in real time, cell-derived exosomes were tagged with green fluorescent protein (GFP)-CD63 while human serum exosomes were rhodamine isothiocynate-labeled. We show that detachment of adherent cells from various substrata induces a rapid and substantial secretion of exosomes, which then concentrate on the cell surfaces and mediate adhesion to various extracellular matrix proteins. We also demonstrate that disruption of lipid rafts with methyl-beta-cyclodextrin (MβCD) inhibits the internalization of exosomes and that annexins are essential for the exosomal uptake mechanisms. Taken together, these data suggest that cellular detachment is accompanied by significant release of exosomes while cellular adhesion and spreading are enhanced by rapid uptake and disposition of exosomes on the cell surface.


Journal of Cellular Biochemistry | 1999

GALECTIN-3 REGULATES THE ADHESIVE INTERACTION BETWEEN BREAST CARCINOMA CELLS AND ELASTIN

Josiah Ochieng; Paula Warfield; Brenda Green-Jarvis; Ian H. Fentie

Galectin‐3 is a beta‐galactoside binding lectin whose precise physiological role is not yet defined. In the present studies, we questioned whether galectin‐3 plays a role in the adhesion of breast carcinoma cells to elastin. The impetus for this analysis was the initial observation that the cellular receptor for elastin, the 67 kDa elastin/laminin protein may have galectin‐like properties (Mecham et al. [1989] J. Biol. Chem. 264:16652–16657). We therefore analyzed the adhesion of breast carcinoma cells to microtiter wells coated with elastin under conditions which eliminate integrin participation in adhesion. The adhesion assay was done in the absence and presence of purified recombinant galectin‐3. We hereby demonstrate that high concentrations of galectin‐3 ligate breast carcinoma cells to microtiter wells coated with elastin. Galectin‐3 also demonstrated a specific binding interaction with purified elastin in a dose and lactose dependent manner. Furthermore we demonstrated by immunoprecipitation that endogenous galectin‐3 in breast carcinoma cells is associated with tropoelastin. Lastly, the breast carcinoma cells which expressed galectin‐3 on their surface, demonstrated enhanced cellular proliferation on elastin compared to galectin‐3 null expressing cells. These studies suggest that galectin‐3 is capable of regulating the interactions between cells and elastin. J. Cell. Biochem. 75:505–514, 1999.


FEBS Letters | 2000

Novel mechanism that Trypanosoma cruzi uses to adhere to the extracellular matrix mediated by human galectin-3.

Tapria N. Moody; Josiah Ochieng; Fernando Villalta

Binding of Trypanosoma cruzi trypomastigotes to laminin is enhanced by galectin‐3, a β‐galactoside binding lectin. The galectin‐3 enhanced binding of trypanosomes to laminin is inhibited by lactose. Co‐immunoprecipitations indicate that galectin‐3 binds to the 45, 32 and 30 kDa trypanosome surface proteins. Binding of galectin‐3 to the 45, 32 and 30 kDa surface proteins is inhibited by lactose. Polyclonal and a monoclonal antibodies to galectin‐3 immunoprecipitated a major 64 kDa trypanosome surface protein. T. cruzi monoclonal antibody to mucin recognized the 45 kDa surface protein. The 45, 32 and 30 kDa surface proteins interact with galectin‐3 in order to enhance trypanosome adhesion to laminin.


Infection and Immunity | 2004

Human Galectin-3 Promotes Trypanosoma cruzi Adhesion to Human Coronary Artery Smooth Muscle Cells

Yuliya Y. Kleshchenko; Tapria N. Moody; Vyacheslav Furtak; Josiah Ochieng; Maria F. Lima; Fernando Villalta

ABSTRACT Human galectin-3 binds to the surface of Trypanosoma cruzi trypomastigotes and human coronary artery smooth muscle (CASM) cells. CASM cells express galectin-3 on their surface and secrete it. Exogenous galectin-3 increased the binding of T. cruzi to CASM cells. Trypanosome binding to CASM cells was enhanced when either T. cruzi or CASM cells were preincubated with galectin-3. Cells stably transfected with galectin-3 antisense show a dramatic decrease in galectin-3 expression and very little T. cruzi adhesion to cells. The addition of galectin-3 to these cells restores their initial capacity to bind to trypanosomes. Thus, host galectin-3 expression is required for T. cruzi adhesion to human cells and exogenous galectin-3 enhances this process, leading to parasite entry.


Experimental Cell Research | 2009

Anchorage-independent growth of breast carcinoma cells is mediated by serum exosomes

Josiah Ochieng; Siddharth Pratap; Atanu K. Khatua; Amos M. Sakwe

We hereby report studies that suggest a role for serum exosomes in the anchorage-independent growth (AIG) of tumor cells. In AIG assays, fetal bovine serum is one of the critical ingredients. We therefore purified exosomes from fetal bovine serum and examined their potential to promote growth of breast carcinoma cells in soft agar and Matrigel after reconstituting them into growth medium (EEM). In all the assays, viable colonies were formed only in the presence of exosomes. Some of the exosomal proteins we identified, have been documented by others and could be considered exosomal markers. Labeled purified exosomes were up-taken by the tumor cells, a process that could be competed out with excess unlabeled vesicles. Our data also suggested that once endocytosed by a cell, the exosomes could be recycled back to the conditioned medium from where they can be up-taken by other cells. We also demonstrated that low concentrations of exosomes activate MAP kinases, suggesting a mechanism by which they maintain the growth of the tumor cells in soft agar. Taken together, our data demonstrate that serum exosomes form a growth promoting platform for AIG of tumor cells and may open a new vista into cancer cell growth in vivo.


Experimental Cell Research | 2011

Annexin A6 contributes to the invasiveness of breast carcinoma cells by influencing the organization and localization of functional focal adhesions

Amos M. Sakwe; Rainelli Koumangoye; Bobby Guillory; Josiah Ochieng

The interaction of annexin A6 (AnxA6) with membrane phospholipids and either specific extracellular matrix (ECM) components or F-actin suggests that it may influence cellular processes associated with rapid plasma membrane reorganization such as cell adhesion and motility. Here, we examined the putative roles of AnxA6 in adhesion-related cellular processes that contribute to breast cancer progression. We show that breast cancer cells secrete annexins via the exosomal pathway and that the secreted annexins are predominantly cell surface-associated. Depletion of AnxA6 in the invasive BT-549 breast cancer cells is accompanied by enhanced anchorage-independent cell growth but cell-cell cohesion, cell adhesion/spreading onto collagen type IV or fetuin-A, cell motility and invasiveness were strongly inhibited. To explain the loss in adhesion/motility, we show that vinculin-based focal adhesions in the AnxA6-depleted BT-549 cells are elongated and randomly distributed. These focal contacts are also functionally defective because the activation of focal adhesion kinase and the phosphoinositide-3 kinase/Akt pathway were strongly inhibited while the MAP kinase pathway remained constitutively active. Compared with normal human breast tissues, reduced AnxA6 expression in breast carcinoma tissues correlates with enhanced cell proliferation. Together this suggests that reduced AnxA6 expression contributes to breast cancer progression by promoting the loss of functional cell-cell and/or cell-ECM contacts and anchorage-independent cell proliferation.


Journal of Biological Chemistry | 2010

Fetuin-A (α2HS-Glycoprotein) Is a Major Serum Adhesive Protein That Mediates Growth Signaling in Breast Tumor Cells

Amos M. Sakwe; Rainelli Koumangoye; Shawn J. Goodwin; Josiah Ochieng

The identity of the cell adhesive factors in fetal bovine serum, commonly used to supplement growth media, remains a mystery due to the plethora of serum proteins. In the present analyses, we showed that fetuin-A, whose function in cellular attachment in tissue culture has been debated for many years, is indeed a major serum cell attachment factor particularly for tumor cells. We are able to report this because of a new purification strategy that has for the first time given us a homogeneous protein band in colloidal Coomassie-stained gels that retains biological activity. The tumor cells adhered to immobilized fetuin-A and not α2-macroglobulin, its major contaminant. The interaction of cells with fetuin-A was driven mainly by Ca2+ ions, and cells growing in regular medium supplemented with fetal bovine serum were just as sensitive to loss of extracellular Ca2+ ions as cells growing in fetuin-A. Fractionation of human serum revealed that cell attachment was confined to the fractions that had fetuin-A. Interestingly, the tumor cells also took up fetuin-A and secreted it back to the medium using an unknown mechanism that can be observed in live cells. The attachment of tumor cells to fetuin-A was accompanied by phosphatidylinositol 3-kinase/Akt activation that was down-regulated in cells that lack annexin-A6, one of the cell surface receptors for fetuin-A. Taken together, our data show the significance of fetuin-A in tumor cell growth mechanisms in vitro and open new research vistas for this protein.


Journal of Biological Chemistry | 2010

Fetuin-A (ahsg) is a major serum adhesive protein that mediates growth signaling in breast tumor cells

Amos M. Sakwe; Rainelli Koumangoye; Shawn J. Goodwin; Josiah Ochieng

The identity of the cell adhesive factors in fetal bovine serum, commonly used to supplement growth media, remains a mystery due to the plethora of serum proteins. In the present analyses, we showed that fetuin-A, whose function in cellular attachment in tissue culture has been debated for many years, is indeed a major serum cell attachment factor particularly for tumor cells. We are able to report this because of a new purification strategy that has for the first time given us a homogeneous protein band in colloidal Coomassie-stained gels that retains biological activity. The tumor cells adhered to immobilized fetuin-A and not α2-macroglobulin, its major contaminant. The interaction of cells with fetuin-A was driven mainly by Ca2+ ions, and cells growing in regular medium supplemented with fetal bovine serum were just as sensitive to loss of extracellular Ca2+ ions as cells growing in fetuin-A. Fractionation of human serum revealed that cell attachment was confined to the fractions that had fetuin-A. Interestingly, the tumor cells also took up fetuin-A and secreted it back to the medium using an unknown mechanism that can be observed in live cells. The attachment of tumor cells to fetuin-A was accompanied by phosphatidylinositol 3-kinase/Akt activation that was down-regulated in cells that lack annexin-A6, one of the cell surface receptors for fetuin-A. Taken together, our data show the significance of fetuin-A in tumor cell growth mechanisms in vitro and open new research vistas for this protein.

Collaboration


Dive into the Josiah Ochieng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge