Next Issue
Volume 3, June
Previous Issue
Volume 2, December
 
 

Biomedicines, Volume 3, Issue 1 (March 2015) – 10 articles , Pages 1-181

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
1248 KiB  
Review
Recent Progress and Advances in HGF/MET-Targeted Therapeutic Agents for Cancer Treatment
by Yilong Zhang, Rajul K. Jain and Min Zhu
Biomedicines 2015, 3(1), 149-181; https://doi.org/10.3390/biomedicines3010149 - 19 Mar 2015
Cited by 23 | Viewed by 10894
Abstract
The hepatocyte growth factor (HGF): MET axis is a ligand-mediated receptor tyrosine kinase pathway that is involved in multiple cellular functions, including proliferation, survival, motility, and morphogenesis. Aberrancy in the HGF/MET pathway has been reported in multiple tumor types and is associated with [...] Read more.
The hepatocyte growth factor (HGF): MET axis is a ligand-mediated receptor tyrosine kinase pathway that is involved in multiple cellular functions, including proliferation, survival, motility, and morphogenesis. Aberrancy in the HGF/MET pathway has been reported in multiple tumor types and is associated with tumor stage and prognosis. Thus, targeting the HGF/MET pathway has become a potential therapeutic strategy in oncology development in the last two decades. A number of novel therapeutic agents—either as therapeutic proteins or small molecules that target the HGF/MET pathway—have been tested in patients with different tumor types in clinical studies. In this review, recent progress in HGF/MET pathway-targeted therapy for cancer treatment, the therapeutic potential of HGF/MET-targeted agents, and challenges in the development of such agents will be discussed. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

758 KiB  
Review
HGF/Met-Signaling Contributes to Immune Regulation by Modulating Tolerogenic and Motogenic Properties of Dendritic Cells
by Jessica Hübel and Thomas Hieronymus
Biomedicines 2015, 3(1), 138-148; https://doi.org/10.3390/biomedicines3010138 - 03 Mar 2015
Cited by 26 | Viewed by 8103
Abstract
Hepatocyte growth factor (HGF)-signaling via Met can induce mitogenic, morphogenic, and motogenic activity in various cell types. Met expression in the immune system is limited to cells with antigen-presenting capacities, including dendritic cells (DCs). Thus, it appears highly conceivable that Met-signaling impacts on [...] Read more.
Hepatocyte growth factor (HGF)-signaling via Met can induce mitogenic, morphogenic, and motogenic activity in various cell types. Met expression in the immune system is limited to cells with antigen-presenting capacities, including dendritic cells (DCs). Thus, it appears highly conceivable that Met-signaling impacts on adaptive immune responses. However, the mechanisms by which HGF imparts its effects on immunological responses are not yet fully understood. DCs possess unique functionalities that are critically involved in controlling both tolerance and immunity. HGF conveys immunoregulatory functions, which strongly correlate with that of DCs orchestrating the apt immune response in inflammation. Therefore, this review focuses on the current knowledge of Met-signaling in DCs with specific emphasis on the morphogenic and motogenic activities. HGF has been identified to play a role in peripheral immune tolerance by directing DC differentiation towards a tolerogenic phenotype. In skin immunity, Met-signaling was shown to drive mobilization of DCs by regulating matrix metalloproteinase activities. This is strikingly reminiscent of the role of Met for regulating a cell fate program during embryonic development, wound healing, and in tumor invasion known as epithelial-mesenchymal transition (EMT). Thus, the concept emerges that an EMT program is executed by Met-signaling in DCs, which will be also discussed. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

1182 KiB  
Article
Anti-Differentiation Effect of Oncogenic Met Receptor in Terminally-Differentiated Myotubes
by Valentina Sala, Simona Gallo, Stefano Gatti, Elisa Vigna, Antonio Ponzetto and Tiziana Crepaldi
Biomedicines 2015, 3(1), 124-137; https://doi.org/10.3390/biomedicines3010124 - 12 Feb 2015
Cited by 3 | Viewed by 7272
Abstract
Activation of the hepatocyte growth factor/Met receptor is involved in muscle regeneration, through promotion of proliferation and inhibition of differentiation in myogenic stem cells (MSCs). We previously described that the specific expression of an oncogenic version of the Met receptor (Tpr–Met) in terminally-differentiated [...] Read more.
Activation of the hepatocyte growth factor/Met receptor is involved in muscle regeneration, through promotion of proliferation and inhibition of differentiation in myogenic stem cells (MSCs). We previously described that the specific expression of an oncogenic version of the Met receptor (Tpr–Met) in terminally-differentiated skeletal muscle causes muscle wasting in vivo. Here, we induced Tpr–Met in differentiated myotube cultures derived from the transgenic mouse. These cultures showed a reduced protein level of myosin heavy chain (MyHC), increased phosphorylation of Erk1,2 MAPK, the formation of giant sacs of myonuclei and the collapse of elongated myotubes. Treatment of the cultures with an inhibitor of the MAPK kinase pathway or with an inhibitor of the proteasome increased the expression levels of MyHC. In addition, the inhibition of the MAPK kinase pathway prevented the formation of myosacs and myotube collapse. Finally, we showed that induction of Tpr–Met in primary myotubes was unable to produce endoreplication in their nuclei. In conclusion, our data indicate that multinucleated, fused myotubes may be forced to disassemble their contractile apparatus by the Tpr–Met oncogenic factor, but they resist the stimulus toward the reactivation of the cell cycle. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

3481 KiB  
Article
Expression of Hepatocyte Growth Factor-Like Protein in Human Wound Tissue and Its Biological Functionality in Human Keratinocytes
by James C. Glasbey, Andrew J. Sanders, David C. Bosanquet, Fiona Ruge, Keith G. Harding and Wen G. Jiang
Biomedicines 2015, 3(1), 110-123; https://doi.org/10.3390/biomedicines3010110 - 04 Feb 2015
Cited by 3 | Viewed by 6142
Abstract
Hepatocyte growth factor-like protein (HGFl) and its receptor, Recepteur d'Origine Nantais (RON), have been implicated in the development of wound chronicity. HGFl and RON expression was detected in acute wound tissue, chronic wound tissue and in normal skin using quantitative polymerase chain reaction [...] Read more.
Hepatocyte growth factor-like protein (HGFl) and its receptor, Recepteur d'Origine Nantais (RON), have been implicated in the development of wound chronicity. HGFl and RON expression was detected in acute wound tissue, chronic wound tissue and in normal skin using quantitative polymerase chain reaction (Q-PCR). HGFl and RON expression was also assessed in chronic healing and chronic non-healing wound tissues using Q-PCR and immunohistochemical staining. Expression was similarly detected in the HaCaT immortalized human keratinocyte cell line using reverse transcription polymerase chain reaction (RT-PCR). rhHGFl was used to assess the impact of this molecule on HaCaT cell functionality using in vitro growth assays and electric cell-substrate impendence sensing (ECIS) migration assays. HGFl and RON transcript expression were significantly increased in acute wound tissue compared to chronic wound tissue and were also elevated, though non-significantly, in comparison to normal skin. Minimal expression was seen in both healing and non-healing chronic wounds. Treatment of HaCaT cells with rhHGFl had no effect on growth rates but did enhance cell migration. This effect was abolished by the addition of a phospholipase C gamma (PLCγ) small molecule inhibitor. The increased expression of HGFl and RON in acute, healing wounds and the pro-migratory effect of HGFl in an in vitro human keratinocyte model, may indicate a role for HGFl in active wound healing. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Figure 1

12481 KiB  
Article
HGF Modulates Actin Cytoskeleton Remodeling and Contraction in Testicular Myoid Cells
by Angela Catizone, Giulia Ricci, Maria Caruso, Michela Galdieri, Katia Corano Scheri, Virginia Di Paolo and Rita Canipari
Biomedicines 2015, 3(1), 89-109; https://doi.org/10.3390/biomedicines3010089 - 28 Jan 2015
Cited by 3 | Viewed by 6202
Abstract
The presence of the HGF/Met system in the testicular myoid cells was first discovered by our group. However, the physiological role of this pathway remains poorly understood. We previously reported that HGF increases uPA secretion and TGF-β activation in cultured tubular fragments and [...] Read more.
The presence of the HGF/Met system in the testicular myoid cells was first discovered by our group. However, the physiological role of this pathway remains poorly understood. We previously reported that HGF increases uPA secretion and TGF-β activation in cultured tubular fragments and that HGF is maximally expressed at Stages VII–VIII of the seminiferous epithelium cycle, when myoid cell contraction occurs. It is well known that the HGF/Met pathway is involved in cytoskeletal remodeling; moreover, the interaction of uPA with its receptor, uPAR, as well as the activation of TGF-β have been reported to be related to the actin cytoskeleton contractility of smooth muscle cells. Herein, we report that HGF induces actin cytoskeleton remodeling in vitro in isolated myoid cells and myoid cell contraction in cultured seminiferous tubules. To better understand these phenomena, we evaluated: (1) the regulation of the uPA machinery in isolated myoid cells after HGF administration; and (2) the effect of uPA or Met inhibition on HGF-treated tubular fragments. Because uPA activates latent TGF-β, the secretion of this factor was also evaluated. We found that both uPA and TGF-β activation increase after HGF administration. In testicular tubular fragments, HGF-induced TGF-β activation and myoid cell contraction are abrogated by uPA or Met inhibitor administration. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

4272 KiB  
Review
HGF/c-MET Axis in Tumor Microenvironment and Metastasis Formation
by Anna Spina, Valeria De Pasquale, Giuliana Cerulo, Pasquale Cocchiaro, Rossella Della Morte, Luigi Avallone and Luigi Michele Pavone
Biomedicines 2015, 3(1), 71-88; https://doi.org/10.3390/biomedicines3010071 - 22 Jan 2015
Cited by 52 | Viewed by 10181
Abstract
Tumor metastases are responsible for approximately 90% of all cancer-related deaths. Metastasis formation is a multistep process that requires acquisition by tumor cells of a malignant phenotype that allows them to escape from the primary tumor site and invade other organs. Each step [...] Read more.
Tumor metastases are responsible for approximately 90% of all cancer-related deaths. Metastasis formation is a multistep process that requires acquisition by tumor cells of a malignant phenotype that allows them to escape from the primary tumor site and invade other organs. Each step of this mechanism involves a deep crosstalk between tumor cells and their microenvironment where the host cells play a key role in influencing metastatic behavior through the release of many secreted factors. Among these signaling molecules, Hepatocyte Growth Factor (HGF) is released by many cell types of the tumor microenvironment to target its receptor c-MET within the cells of the primary tumor. Many studies reveal that HGF/c-MET axis is implicated in various human cancers, and genetic and epigenetic gain of functions of this signaling contributes to cancer development through a variety of mechanisms. In this review, we describe the specific types of cells in the tumor microenvironment that release HGF in order to promote the metastatic outgrowth through the activation of extracellular matrix remodeling, inflammation, migration, angiogenesis, and invasion. We dissect the potential use of new molecules that interfere with the HGF/c-MET axis as therapeutic targets for future clinical trials in cancer disease. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

3677 KiB  
Review
c-Met and Other Cell Surface Molecules: Interaction, Activation and Functional Consequences
by Giuditta Viticchiè and Patricia A. J. Muller
Biomedicines 2015, 3(1), 46-70; https://doi.org/10.3390/biomedicines3010046 - 15 Jan 2015
Cited by 58 | Viewed by 11456
Abstract
The c-Met receptor, also known as the HGF receptor, is one of the most studied tyrosine kinase receptors, yet its biological functions and activation mechanisms are still not fully understood. c-Met has been implicated in embryonic development and organogenesis, in tissue remodelling homeostasis [...] Read more.
The c-Met receptor, also known as the HGF receptor, is one of the most studied tyrosine kinase receptors, yet its biological functions and activation mechanisms are still not fully understood. c-Met has been implicated in embryonic development and organogenesis, in tissue remodelling homeostasis and repair and in cancer metastasis. These functions are indicative of the many cellular processes in which the receptor plays a role, including cell motility, scattering, survival and proliferation. In the context of malignancy, sustained activation of c-Met leads to a signalling cascade involving a multitude of kinases that initiate an invasive and metastatic program. Many proteins can affect the activation of c-Met, including a variety of other cell surface and membrane-spanning molecules or receptors. Some cell surface molecules share structural homology with the c-Met extracellular domain and can activate c-Met via clustering through this domain (e.g., plexins), whereas other receptor tyrosine kinases can enhance c-Met activation and signalling through intracellular signalling cascades (e.g., EGFR). In this review, we provide an overview of c-Met interactions and crosstalk with partner molecules and the functional consequences of these interactions on c-Met activation and downstream signalling, c-Met intracellular localization/recycling and c-Met degradation. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

582 KiB  
Editorial
Acknowledgement to Reviewers of Biomedicines in 2014
by Biomedicines Editorial Office
Biomedicines 2015, 3(1), 45; https://doi.org/10.3390/biomedicines3010045 - 09 Jan 2015
Viewed by 3800
Abstract
The editors of Biomedicines would like to express their sincere gratitude to the following reviewers for assessing manuscripts in 2014:[...] Full article
857 KiB  
Review
c-Met and miRs in Cancer
by Simona Giglio and Andrea Vecchione
Biomedicines 2015, 3(1), 32-44; https://doi.org/10.3390/biomedicines3010032 - 05 Jan 2015
Cited by 12 | Viewed by 7445
Abstract
c-Met, a member of the receptor tyrosine kinase family, is involved in a wide range of cellular processes, including tumor survival, cell growth, angiogenesis and metastasis, and resulting in overexpression in many human cancers, leading to a constitutive activation of the downstream pathways. [...] Read more.
c-Met, a member of the receptor tyrosine kinase family, is involved in a wide range of cellular processes, including tumor survival, cell growth, angiogenesis and metastasis, and resulting in overexpression in many human cancers, leading to a constitutive activation of the downstream pathways. Recently identified MicroRNAs are a family of small noncoding RNA molecules, extensively studied in cancer, that exert their action by inhibiting gene expression at the posttranscriptional level in several biological processes. Aberrant regulation of microRNAs expression has been implicated in the pathogenesis of different human neoplasia. Several publications point out the connections between c-Met and its ligand hepatocyte growth factor (HGF) and microRNAs. This review summarizes the current knowledge about the interplay between c-Met/HGF and microRNAs and provides evidence that microRNAs are a novel and additional system to regulate c-Met expression in tumors. In the future, microRNAs connected to c-Met may provide an additional option to inhibiting this oncogene from orchestrating an invasive growth program. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Graphical abstract

7075 KiB  
Review
Physiological Signaling and Structure of the HGF Receptor MET
by Gianluca Baldanzi and Andrea Graziani
Biomedicines 2015, 3(1), 1-31; https://doi.org/10.3390/biomedicines3010001 - 31 Dec 2014
Cited by 35 | Viewed by 10065
Abstract
The “hepatocyte growth factor” also known as “scatter factor”, is a multifunctional cytokine with the peculiar ability of simultaneously triggering epithelial cell proliferation, movement and survival. The combination of those proprieties results in the induction of an epithelial to mesenchymal transition in target [...] Read more.
The “hepatocyte growth factor” also known as “scatter factor”, is a multifunctional cytokine with the peculiar ability of simultaneously triggering epithelial cell proliferation, movement and survival. The combination of those proprieties results in the induction of an epithelial to mesenchymal transition in target cells, fundamental for embryogenesis but also exploited by tumor cells during metastatization. The hepatocyte growth factor receptor, MET, is a proto-oncogene and a prototypical transmembrane tyrosine kinase receptor. Inhere we discuss the MET molecular structure and the hepatocyte growth factor driven physiological signaling which coordinates epithelial proliferation, motility and morphogenesis. Full article
(This article belongs to the Special Issue New aspects of the Hepatocyte Growth Factor/c-Met System)
Show Figures

Figure 1

Previous Issue
Next Issue
Back to TopTop