[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (2)

Search Parameters:
Keywords = liver fibrocarcinogenesis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1227 KiB  
Review
Clinico-Pathological Importance of TGF-β/Phospho-Smad Signaling during Human Hepatic Fibrocarcinogenesis
by Katsunori Yoshida, Koichi Matsuzaki, Miki Murata, Takashi Yamaguchi, Kanehiko Suwa and Kazuichi Okazaki
Cancers 2018, 10(6), 183; https://doi.org/10.3390/cancers10060183 - 5 Jun 2018
Cited by 64 | Viewed by 8351
Abstract
Chronic viral hepatitis is a global public health problem, with approximately 570 million persons chronically infected. Hepatitis B and C viruses increase the risk of morbidity and mortality from liver cirrhosis, hepatocellular carcinoma (HCC), and extrahepatic complications that develop. Hepatitis virus infection induces [...] Read more.
Chronic viral hepatitis is a global public health problem, with approximately 570 million persons chronically infected. Hepatitis B and C viruses increase the risk of morbidity and mortality from liver cirrhosis, hepatocellular carcinoma (HCC), and extrahepatic complications that develop. Hepatitis virus infection induces transforming growth factor (TGF)-β, which influences microenvironments within the infected liver. TGF-β promotes liver fibrosis by up-regulating extracellular matrix production by hepatic stellate cells. TGF-β is also up-regulated in patients with HCC, in whom it contributes importantly to bringing about a favorable microenvironment for tumor growth. Thus, TGF-β is thought to be a major factor regulating liver fibrosis and carcinogenesis. Since TGF-β carries out regulatory signaling by influencing the phosphorylation of Smads, we have generated several kinds of phospho-specific antibodies to Smad2/3. Using these, we have identified three types of phospohorylated forms: COOH-terminally phosphorylated Smad2/3 (pSmad2C and pSmad3C), linker phosphorylated Smad2/3 (pSmad2L and pSmad3L), and dually phosphorylated Smad3 (pSmad2L/C and pSmad3L/C). TGF-β-mediated pSmad2/3C signaling terminates cell proliferation; on the other hand, cytokine-induced pSmad3L signaling accelerates cell proliferation and promotes fibrogenesis. This review addresses TGF-β/Smad signal transduction in chronic liver injuries and carcinogenic processes. We also discuss the reversibility of Smad signaling after antiviral therapy. Full article
(This article belongs to the Special Issue TGF-Beta Signaling in Cancer)
Show Figures

Figure 1

Figure 1
<p>Differential phospho-Smad signals between tumor suppression and fibrocarcinogenesis. (<b>A</b>) Activated transforming growth factor (TGF)-β type I receptor (TβRI) phosphorylates COOH-tail serine residues of Smad2 and Smad3. Both COOH-terminally phosphorylated Smad2/3 (pSmad2C and pSmad3C) translocate with Smad4 to the nuclei of quiescent hepatocytes after regeneration. Smad2/3/4, complex binds the p21<sup>waf1</sup> promoter and suppresses cell growth; (<b>B</b>) Pro-inflammatory cytokines (CK) such as tumor necrosis factor-α activate c-Jun N-terminal kinase (JNK), which phosphorylates the linker regions of Smad2 and Smad3. Linker phosphorylated Smad3 (PSmad3L) translocates with Smad4 to the nucleus and binds plasminogen activator inhibitor type 1 (PAI-1) promoter. Linker phosphorylated Smad2 (PSmad2L) is localized in the cytoplasm, and Smad2 translocates to the nucleus only after COOH-tail phosphorylation by TβRI. PSmad2L/C in cooperation with pSmad3L and Smad4 stimulate PAI-1 transcription and extracellular matrix (ECM) deposition. PSmad3L up-regulates c-Myc and stimulates cell growth, while suppressing the pSmad3C-mediated tumor suppressive pathway.</p>
Full article ">Figure 2
<p>Phenotypic alternations of hepatocytes and HSC during the fibrocarcinogenic process in human chronic liver diseases. Quiescent hepatic stellate cells (HSC) are characterized by retinoid droplets in the cytoplasm and maintain liver homeostasis. HSC undergo constitutive activation to become myofibroblasts (MFB)-like cells after liver injury. MFB persistently produce an extracellular matrix (ECM) and induce liver fibrosis. The contraction of MFB contributes to increased portal resistance during liver fibrosis that presumably is reversible until the thickened septae, intrahepatic shunts, and lobular distortion that are characteristic of cirrhosis development, leading to fixed increases in portal pressure. Chronic liver damage promotes recurrent cycles of cellular proliferation, inflammation, fibrosis, and carcinogenesis. In pre-neoplastic hepatocytes, several growth factors and cytokines activate proliferation and invasion. As human hepatitis virus-related chronic liver diseases progress, chronic inflammation and hepatitis virus additively accelerate liver fibrosis and increase the risk of hepatocellular carcinoma (HCC). Genetic and epigenetic changes in the liver result in carcinogenesis. Effective antiviral therapy can reverse the pre-neoplastic properties of hepatocytes to a tumor-suppressive mode before the occurrence of genetic mutations that have been implicated for HCC occurrence.</p>
Full article ">
1553 KiB  
Review
Reversible Human TGF-β Signal Shifting between Tumor Suppression and Fibro-Carcinogenesis: Implications of Smad Phospho-Isoforms for Hepatic Epithelial-Mesenchymal Transitions
by Katsunori Yoshida, Miki Murata, Takashi Yamaguchi, Koichi Matsuzaki and Kazuichi Okazaki
J. Clin. Med. 2016, 5(1), 7; https://doi.org/10.3390/jcm5010007 - 12 Jan 2016
Cited by 40 | Viewed by 7657
Abstract
Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are observed during both physiological liver wound healing and the pathological fibrotic/carcinogenic (fibro-carcinogenetic) process. TGF-β and pro-inflammatory cytokine are considered to be the major factors accelerating liver fibrosis and promoting liver carcinogenesis. Smads, consisting of intermediate [...] Read more.
Epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are observed during both physiological liver wound healing and the pathological fibrotic/carcinogenic (fibro-carcinogenetic) process. TGF-β and pro-inflammatory cytokine are considered to be the major factors accelerating liver fibrosis and promoting liver carcinogenesis. Smads, consisting of intermediate linker regions connecting Mad homology domains, act as the intracellular mediators of the TGF-β signal transduction pathway. As the TGF-β receptors, c-Jun N-terminal kinase and cyclin-dependent kinase, differentially phosphorylate Smad2/3, we have generated numerous antibodies against linker (L) and C-terminal (C) phosphorylation sites in Smad2/3 and identified four types of phosphorylated forms: cytostatic COOH-terminally-phosphorylated Smad3 (pSmad3C), mitogenic pSmad3L (Ser-213) signaling, fibrogenic pSmad2L (Ser-245/250/255)/C signaling and migratory pSmad2/3L (Thr-220/179)/C signaling. After acute liver injury, TGF-β upregulates pSmad3C signaling and terminates pSmad3L (Ser-213)-mediated hepatocyte proliferation. TGF-β and pro-inflammatory cytokines cooperatively enhance collagen synthesis by upregulating pSmad2L (Thr-220)/C and pSmad3L (Thr-179)/C pathways in activated hepatic stellate cells. During chronic liver injuries, hepatocytes persistently affected by TGF-β and pro-inflammatory cytokines eventually become pre-neoplastic hepatocytes. Both myofibroblasts and pre-neoplastic hepatocyte exhibit the same carcinogenic (mitogenic) pSmad3L (Ser-213) and fibrogenic pSmad2L (Ser-245/250/255)/C signaling, with acquisition of fibro-carcinogenic properties and increasing risk of hepatocellular carcinoma (HCC). Firstly, we review phospho-Smad-isoform signalings in epithelial and mesenchymal cells in physiological and pathological conditions and then consider Smad linker phosphorylation as a potential target for pathological EMT during human fibro-carcinogenesis, because human Smad phospho-isoform signals can reverse from fibro-carcinogenesis to tumor-suppression in a process of MET after therapy. Full article
(This article belongs to the Special Issue Epithelial-Mesenchymal Transition)
Show Figures

Figure 1

Figure 1
<p>Liver regeneration-related EMT and phospho-Smad signaling in acute liver disease. (<b>A</b>) Quiescent hepatic stellate cells (HSC) are characterized by retinoid droplets in the cytoplasm. Acute liver injury caused HSC activation and hepatocyte damage, necrosis and EMT. Activated HSC move from the space of Disse to sites of damage where the activated HSC contribute to tissue repair by producing large amounts of collagen. HSC also play an important role in secreting TGF-β. (<b>B</b>) Catalytically-active TβRI phosphorylates COOH-tail serine residues of Smad2 and Smad3. Both pSmad2C and pSmad3C are localized in the nuclei of hepatocytes and mesenchymal cells in acute injured liver. After binding with Smad4, pSmad2/3C translocate with Smad4 to the nucleus and bind to the collagen promoter. pSmad2/3C stimulate extracellular matrix (ECM) deposition and suppress cell growth by c-Myc inhibition. However, Smad7 induced by the pSmad3L/C signal terminates the fibrogenic phospho-Smad signaling. This negative feedback mechanism of the fibrogenic TGF-β/CK signal results in a transient collagen synthesis in the activated HSC, which may thus contribute to tissue repair.</p>
Full article ">Figure 2
<p>Liver fibro-carcinogenesis-related EMT and phospho-Smad signaling during chronic liver disease. (<b>A</b>) Prolonged exposure to chronic injury; HSC undergo constitutive activation to become myofibroblasts (MFB)-like cells, which persistently induce deposition of ECM and liver fibrosis. Continuous insults will shift EMT-like cells to complete EMT and pre-neoplastic hepatocytes. (<b>B</b>) During chronic liver injury, pro-inflammatory cytokines (CK), such as TNF-α activate JNK, result in phosphorylation of both Smad2L and Smad3L, both in MFB and pre-neoplastic hepatocyte. P-Smad3L translocates with Smad4 to the nucleus and binds the PAI-1 promoter. After COOH-tail phosphorylation of cytoplasmic pSmad2L by TβRI, pSmad2L/C translocates to the nucleus. Both pSmad2L/C and pSmad3L stimulates PAI-1 transcription and ECM deposition, while they suppress the pSmad3C-mediated tumor suppressive pathway. Pre-neoplastic hepatocytes exhibit the same oncogenic (mitogenic) pSmad3L and fibrogenic pSmad2L signaling as MFB, thereby accelerating liver fibrosis and increasing the risk of HCC. In contrast to Smad7 induction in HSC via the pSmad3C pathway, pSmad3L cannot induce Smad7 in MFB and pre-neoplastic hepatocytes (left). Under a low level of Smad7, the fibrogenic phospho-Smad signaling can constitutively promote ECM deposition by MFB, which may eventually develop into accelerated liver fibro-carcinogenesis.</p>
Full article ">Figure 3
<p>Phosphorylated Smad2/3 signaling fibro-carcinogenesis. As human hepatitis virus-related chronic liver diseases progress, chronic inflammation and hepatitis virus additively shift hepatocytic Smad phospho-isoform signaling from tumor-suppressive pSmad3C to the fibro-carcinogenic pSmad3L and pSmad2L/C pathway. Anti-viral therapy can reverse phospho-Smad signaling from fibro-carcinogenesis to tumor suppression. Type 2 EMT promotes liver fibrosis induced by chronic inflammation. Type 3 EMT exacerbates the HCC phenotype by upregulating invasive and metastatic potential.</p>
Full article ">
Back to TopTop