1
|
de Haan LR, van Golen RF, Heger M. Molecular Pathways Governing the Termination of Liver Regeneration. Pharmacol Rev 2024; 76:500-558. [PMID: 38697856 DOI: 10.1124/pharmrev.123.000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 05/05/2024] Open
Abstract
The liver has the unique capacity to regenerate, and up to 70% of the liver can be removed without detrimental consequences to the organism. Liver regeneration is a complex process involving multiple signaling networks and organs. Liver regeneration proceeds through three phases: the initiation phase, the growth phase, and the termination phase. Termination of liver regeneration occurs when the liver reaches a liver-to-body weight that is required for homeostasis, the so-called "hepatostat." The initiation and growth phases have been the subject of many studies. The molecular pathways that govern the termination phase, however, remain to be fully elucidated. This review summarizes the pathways and molecules that signal the cessation of liver regrowth after partial hepatectomy and answers the question, "What factors drive the hepatostat?" SIGNIFICANCE STATEMENT: Unraveling the pathways underlying the cessation of liver regeneration enables the identification of druggable targets that will allow us to gain pharmacological control over liver regeneration. For these purposes, it would be useful to understand why the regenerative capacity of the liver is hampered under certain pathological circumstances so as to artificially modulate the regenerative processes (e.g., by blocking the cessation pathways) to improve clinical outcomes and safeguard the patient's life.
Collapse
Affiliation(s)
- Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Rowan F van Golen
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| |
Collapse
|
2
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Zhai Y, Ye SY, Wang QS, Xiong RP, Fu SY, Du H, Xu YW, Peng Y, Huang ZZ, Yang N, Zhao Y, Ning YL, Li P, Zhou YG. Overexpressed ski efficiently promotes neurorestoration, increases neuronal regeneration, and reduces astrogliosis after traumatic brain injury. Gene Ther 2023; 30:75-87. [PMID: 35132206 DOI: 10.1038/s41434-022-00320-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 12/31/2021] [Accepted: 01/20/2022] [Indexed: 11/09/2022]
Abstract
Traumatic brain injury (TBI) survivors suffer from long-term disability and neuropsychiatric sequelae due to irreparable brain tissue destruction. However, there are still few efficient therapies to promote neurorestoration in damaged brain tissue. This study aimed to investigate whether the pro-oncogenic gene ski can promote neurorestoration after TBI. We established a ski-overexpressing experimental TBI mouse model using adenovirus-mediated overexpression through immediate injection after injury. Hematoxylin-eosin staining, MRI-based 3D lesion volume reconstruction, neurobehavioral tests, and analyses of neuronal regeneration and astrogliosis were used to assess neurorestorative efficiency. The effects of ski overexpression on the proliferation of cultured immature neurons and astrocytes were evaluated using imaging flow cytometry. The Ski protein level increased in the perilesional region at 3 days post injury. ski overexpression further elevated Ski protein levels up to 14 days post injury. Lesion volume was attenuated by approximately 36-55% after ski overexpression, with better neurobehavioral recovery, more newborn immature and mature neurons, and less astrogliosis in the perilesional region. Imaging flow cytometry results showed that ski overexpression elevated the proliferation rate of immature neurons and reduced the proliferation rate of astrocytes. These results show that ski can be considered a novel neurorestoration-related gene that effectively promotes neurorestoration, facilitates neuronal regeneration, and reduces astrogliosis after TBI.
Collapse
Affiliation(s)
- Yu Zhai
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Shi-Yang Ye
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Qiu-Shi Wang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.,Department of Pathology, Research Institute of Surgery and Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ren-Ping Xiong
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Sheng-Yu Fu
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Hao Du
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ya-Wei Xu
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Yan Peng
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Zhi-Zhong Huang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Nan Yang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Yan Zhao
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ya-Lei Ning
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ping Li
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.
| | - Yuan-Guo Zhou
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
4
|
Liao HY, Ran R, Da CM, Wang ZQ, Zhou KS, Zhang HH. Ski Regulates the Inflammatory Response of Reactive Astrocytes Induced by Oxygen Glucose Deprivation/Reoxygenation (OGD/R) Through the NF-κB Pathway. Neuroscience 2022; 490:250-263. [PMID: 35339646 DOI: 10.1016/j.neuroscience.2022.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 10/18/2022]
Abstract
Spinal cord injury (SCI) is a common disease of the nervous system, including primary and secondary injuries. Neuronal inflammation after SCI is the most important pathological process of SCI and a chemical barrier to nerve function recovery after injury. Ski, an evolutionarily conserved functional transcriptional regulator protein, is upregulated in reactive astrocytes after SCI and regulates the biological characteristics of astrocytes. However, its role in the glial inflammatory response triggered by reactive astrocytes after spinal cord ischemia and its exact mechanism remains unclear. This study investigated the role and mechanism of Ski in the inflammatory response triggered by reactive astrocytes induced by oxygen and sugar deprivation/reoxygenation (OGD/R) model in vitro. In the ODG/R model, Ski expression was upregulated. In contrast, Ski upregulation was accompanied by increased levels of iNOS, IL-1β, IL-6, TNF-α, and other inflammation-related factors. These results indicated that the inflammatory response triggered by astrocytes was significantly enhanced in OGD/R-stimulated astrocytes. Astrocytes were transfected with Ski specific siRNA to knock out Ski and subsequently attenuate OGD-induced astrocyte-triggered inflammation. Our results also suggest that Ski downregulation downregulates the expression of iNOS, IL-1β, IL-6, and TNF-α in OGD/R-induced reactive astrocytes by inhibiting the activity of the NF-κB signaling pathway. In conclusion, downregulation of Ski can effectively inhibit glial inflammation in SCI by inhibiting the activity of the NF-κB pathway. These findings suggest that Ski is a promising therapeutic target for inflammatory responses after SCI.In conclusion, Ski downregulation can effectively inhibit glial inflammation in SCI by inhibiting the activity of the NF-κB pathway. These findings suggest that Ski might serve as a promising target for the treatment of inflammatory responses after SCI.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Rui Ran
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Chao-Ming Da
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China; Gansu Provincial Maternal and Child Health Hospital, 143 Qilihe North Street, Lanzhou 730050, PR China
| | - Zhi-Qiang Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Kai-Sheng Zhou
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
5
|
García IC, Villalba JS, Iovino D, Franchi C, Iori V, Pettinato G, Inversini D, Amico F, Ietto G. Liver Trauma: Until When We Have to Delay Surgery? A Review. Life (Basel) 2022; 12:life12050694. [PMID: 35629360 PMCID: PMC9143295 DOI: 10.3390/life12050694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 01/09/2023] Open
Abstract
Liver involvement after abdominal blunt trauma must be expected, and in up to 30% of cases, spleen, kidney, and pancreas injuries may coexist. Whenever hemodynamics conditions do not contraindicate the overcoming of the ancient dogma according to which exploratory laparotomy should be performed after every major abdominal trauma, a CT scan has to clarify the liver lesions so as to determine the optimal management strategy. Except for complete vascular avulsion, no liver trauma grade precludes nonoperative management. Every attempt to treat the injured liver by avoiding a strong surgical approach may be considered. Each time, a nonoperative management (NOM) consisting of a basic “wait and see” attitude combined with systemic support and blood replacement are inadequate. Embolization should be considered to stop the bleeding. Percutaneous drainage of collections, endoscopic retrograde cholangiopancreatography (ERCP) with papilla sphincterotomy or stent placement and percutaneous transhepatic biliary drainage (PTBD) may avoid, or at least delay, surgical reconstruction or resection until systemic and hepatic inflammatory remodeling are resolved. The pathophysiological principle sustaining these leanings is based on the opportunity to limit the further release of cell debris fragments acting as damage-associated molecular patterns (DAMPs) and the following stress response associated with the consequent immune suppression after trauma. The main goal will be a faster recovery combined with limited cell death of the liver through the ischemic events that may directly follow the trauma, exacerbated by hemostatic procedures and surgery, in order to reduce the gross distortion of a regenerated liver.
Collapse
Affiliation(s)
- Inés Cañas García
- General and Digestive Surgery, Hospital Clínico San Cecilio of Granada, 18002 Granada, Spain;
| | - Julio Santoyo Villalba
- General and Digestive Surgery, Hospital Virgen de Las Nieves of Granada, 18002 Granada, Spain;
| | - Domenico Iovino
- General, Emergency and Transplant Surgery Department, ASST-Settelaghi and University of Insubria, 21100 Varese, Italy; (D.I.); (C.F.); (V.I.); (D.I.)
| | - Caterina Franchi
- General, Emergency and Transplant Surgery Department, ASST-Settelaghi and University of Insubria, 21100 Varese, Italy; (D.I.); (C.F.); (V.I.); (D.I.)
| | - Valentina Iori
- General, Emergency and Transplant Surgery Department, ASST-Settelaghi and University of Insubria, 21100 Varese, Italy; (D.I.); (C.F.); (V.I.); (D.I.)
| | - Giuseppe Pettinato
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Davide Inversini
- General, Emergency and Transplant Surgery Department, ASST-Settelaghi and University of Insubria, 21100 Varese, Italy; (D.I.); (C.F.); (V.I.); (D.I.)
| | - Francesco Amico
- Trauma Service, Department of Surgery, University of Newcastle, Newcastle 2308, Australia;
| | - Giuseppe Ietto
- General, Emergency and Transplant Surgery Department, ASST-Settelaghi and University of Insubria, 21100 Varese, Italy; (D.I.); (C.F.); (V.I.); (D.I.)
- Correspondence: ; Tel.: +39-339-8758024
| |
Collapse
|
6
|
Zuñiga-Aguilar E, Ramírez-Fernández O. Fibrosis and hepatic regeneration mechanism. Transl Gastroenterol Hepatol 2022; 7:9. [PMID: 35243118 PMCID: PMC8826211 DOI: 10.21037/tgh.2020.02.21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/10/2020] [Indexed: 11/26/2023] Open
Abstract
Liver cirrhosis is the final stage of continuous hepatic inflammatory activity derived by viral, metabolic or autoimmune origin. In the last years, cirrhosis was considered a unique and static condition; recently was accepted some patients subgroups with different liver injury degrees that coexist under the same diagnosis, with implications about the natural disease history. The liver growth factor (LGF) is a potent in vivo and in vitro mitogenic agent and an inducer of hepatic regeneration (HR) through the hepatocytes DNA synthesis. The clinical implications of the LGF levels in cirrhosis, are not clear and even with having a fundamental role in the liver regeneration processes, the studies suggest that it could be a cirrhosis severity marker, in acute liver failure and in chronic hepatitis. Its role as predictor of mortality in fulminant hepatic insufficiency patients has been suggested. HR is one of the most enigmatic and fascinating biological phenomena. The rapid volume and liver function restoration after a major hepatectomy (>70%) or severe hepatocellular damage and its strict regulation of tissue damage response after the cessation, is an exclusive property of the liver. HR is the clinical applications fundament, such as extensive hepatic resections (>70% of the liver parenchyma), segmental transplantation or living donor transplantation, sequential hepatectomies, isolated portal embolization or associated with in situ hepatic transection, temporary artificial support in acute liver failure and the possible cell therapy clinical applications.
Collapse
Affiliation(s)
- Esmeralda Zuñiga-Aguilar
- Universidad Autonoma de Ciudad Juárez, Depto de Ingeniería Eléctrica y Computación, Ciudad Juárez, Chih., México
| | - Odin Ramírez-Fernández
- Tecnologico Nacional de Mexico, Depto. De Ciencias Basicas, Tlalnepantla de Baz, Mexico
- Facultad de Medicina, HIPAM, Universidad Nacional Autonoma de Mexico, Ciudad de México, Mexico
| |
Collapse
|
7
|
Liao HY, Wang ZQ, Da CM, Zhou KS, Zhang HH. Ski regulates proliferation and migration of reactive astrocytes induced by lipopolysaccharide (LPS) through PI3K/Akt pathway. J Neuroimmunol 2022; 364:577807. [PMID: 35007896 DOI: 10.1016/j.jneuroim.2022.577807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a leading cause of disability and death worldwide. Reactive astrogliosis, a typical feature of SCI, undergoes various molecular and morphological changes and contributes to glial scar formation, which impedes axonal regeneration. Ski is a novel molecule that regulates the biological characteristics of astrocytes after spinal cord injury, but its function and the exact mechanism of its overexpression in reactive astrocyte proliferation and migration after SCI remain unclear. The purpose of this study was to elucidate the effect and mechanism of Ski on the proliferation and migration of reactive astrocytes, and to regulate the spatiotemporal formation of glial scars after SCI. In an in vitro lipopolysaccharide (LPS)-induced astrocyte injury model, the expression of Ski was upregulated in a time-dependent manner in LPS-induced astrocytes, and the upregulation of Ski was accompanied by that of PCNA, CDK4, CyclinD1, and other proliferation-related proteins. Our findings suggest that Ski promotes the proliferation and migration of reactive astrocytes. Next, astrocytes were transfected with a specific lentivirus to cause the overexpression of Ski, which significantly enhanced the proliferation and migration of reactive astrocytes and LPS-induced activation of the PI3K/Akt pathway. The PI3K/Akt pathway inhibitor LY294002 significantly inhibited the proliferation and migration of LPS-induced reactive astrocytes after Ski overexpression. In conclusion, Ski regulates LPS-induced astrocyte proliferation and migration through the PI3K/Akt pathway, making Ski a promising target for strategies to combat glial scarring after SCI.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Zhi-Qiang Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Chao-Ming Da
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Kai-Sheng Zhou
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
8
|
Xu F, Tautenhahn HM, Dirsch O, Dahmen U. Modulation of Autophagy: A Novel "Rejuvenation" Strategy for the Aging Liver. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6611126. [PMID: 33628363 PMCID: PMC7889356 DOI: 10.1155/2021/6611126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/08/2020] [Accepted: 01/23/2021] [Indexed: 12/11/2022]
Abstract
Aging is a natural life process which leads to a gradual decline of essential physiological processes. For the liver, it leads to alterations in histomorphology (steatosis and fibrosis) and function (protein synthesis and energy generation) and affects central hepatocellular processes (autophagy, mitochondrial respiration, and hepatocyte proliferation). These alterations do not only impair the metabolic capacity of the liver but also represent important factors in the pathogenesis of malignant liver disease. Autophagy is a recycling process for eukaryotic cells to degrade dysfunctional intracellular components and to reuse the basic substances. It plays a crucial role in maintaining cell homeostasis and in resisting environmental stress. Emerging evidence shows that modulating autophagy seems to be effective in improving the age-related alterations of the liver. However, autophagy is a double-edged sword for the aged liver. Upregulating autophagy alleviates hepatic steatosis and ROS-induced cellular stress and promotes hepatocyte proliferation but may aggravate hepatic fibrosis. Therefore, a well-balanced autophagy modulation strategy might be suitable to alleviate age-related liver dysfunction. Conclusion. Modulation of autophagy is a promising strategy for "rejuvenation" of the aged liver. Detailed knowledge regarding the most devastating processes in the individual patient is needed to effectively counteract aging of the liver without causing obvious harm.
Collapse
Affiliation(s)
- Fengming Xu
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena 07747, Germany
| | - Hans-Michael Tautenhahn
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena 07747, Germany
| | - Olaf Dirsch
- Institute of Pathology, Klinikum Chemnitz gGmbH, Chemnitz 09111, Germany
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena 07747, Germany
| |
Collapse
|
9
|
Abarca-Buis RF, Mandujano-Tinoco EA, Cabrera-Wrooman A, Krötzsch E. The complexity of TGFβ/activin signaling in regeneration. J Cell Commun Signal 2021; 15:7-23. [PMID: 33481173 DOI: 10.1007/s12079-021-00605-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
The role of transforming growth factor β TGFβ/activin signaling in wound repair and regeneration is highly conserved in the animal kingdom. Various studies have shown that TGF-β/activin signaling can either promote or inhibit different aspects of the regeneration process (i.e., proliferation, differentiation, and re-epithelialization). It has been demonstrated in several biological systems that some of the different cellular responses promoted by TGFβ/activin signaling depend on the activation of Smad-dependent or Smad-independent signal transduction pathways. In the context of regeneration and wound healing, it has been shown that the type of R-Smad stimulated determines the different effects that can be obtained. However, neither the possible roles of Smad-independent pathways nor the interaction of the TGFβ/activin pathway with other complex signaling networks involved in the regenerative process has been studied extensively. Here, we review the important aspects concerning the TGFβ/activin signaling pathway in the regeneration process. We discuss data regarding the role of TGF-β/activin in the most common animal regenerative models to demonstrate how this signaling promotes or inhibits regeneration, depending on the cellular context.
Collapse
Affiliation(s)
- René Fernando Abarca-Buis
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico.
| | - Edna Ayerim Mandujano-Tinoco
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Alejandro Cabrera-Wrooman
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| | - Edgar Krötzsch
- Laboratory of Connective Tissue, Centro Nacional de Investigación y Atención de Quemados, Instituto Nacional de Rehabilitación "Luís Guillermo Ibarra Ibarra", Calzada México-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, 14389, Mexico City, Mexico
| |
Collapse
|
10
|
Liao HY, Da CM, Wu ZL, Zhang HH. Ski: Double roles in cancers. Clin Biochem 2020; 87:1-12. [PMID: 33188772 DOI: 10.1016/j.clinbiochem.2020.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
The Ski (Sloan-Kettering Institute) is an evolutionarily conserved protein that plays a dual role as an oncoprotein and tumor suppressor gene in the development of human cancer. The Ski oncogene was first identified as a transforming protein of the avian Sloan-Kettering retrovirus in 1986. Since its discovery, Ski has been identified as a carcinogenic regulator in a variety of malignant tumors. Later, it was reported that Ski regulates the occurrence and development of some cancers by acting as an oncogene. Ski mediates the proliferation, differentiation, metastasis, and invasion of numerous cancer cells through various mechanisms. Several studies have shown that Ski expression is correlated with the clinical characteristics of cancer patients and is a promising biomarker and therapeutic target for cancer. In this review, we summarize the mechanisms and potential clinical implications of Ski in dimorphism, cancer occurrence, and progression in various types of cancer.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Chao-Ming Da
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Zuo-Long Wu
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| |
Collapse
|
11
|
Ríos-López DG, Aranda-López Y, Sosa-Garrocho M, Macías-Silva M. La plasticidad del hepatocito y su relevancia en la fisiología y la patología hepática. TIP REVISTA ESPECIALIZADA EN CIENCIAS QUÍMICO-BIOLÓGICAS 2020. [DOI: 10.22201/fesz.23958723e.2020.0.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
El hígado es uno de los principales órganos encargados de mantener la homeostasis en vertebrados, además de poseer una gran capacidad regenerativa. El hígado está constituido por diversos tipos celulares que de forma coordinada contribuyen para que el órgano funcione eficientemente. Los hepatocitos representan el tipo celular principal de este órgano y llevan a cabo la mayoría de sus actividades; además, constituyen una población heterogénea de células epiteliales con funciones especializadas en el metabolismo. El fenotipo de los hepatocitos está controlado por diferentes vías de señalización, como la vía del TGFβ/Smads, la ruta Hippo/YAP-TAZ y la vía Wnt/β-catenina, entre otras. Los hepatocitos son células que se encuentran normalmente en un estado quiescente, aunque cuentan con una plasticidad intrínseca que se manifiesta en respuesta a diversos daños en el hígado; así, estas células reactivan su capacidad proliferativa o cambian su fenotipo a través de procesos celulares como la transdiferenciación o la transformación, para contribuir a mantener la homeostasis del órgano en condiciones saludables o desarrollar diversas patologías.
Collapse
|
12
|
Xu F, Hua C, Tautenhahn HM, Dirsch O, Dahmen U. The Role of Autophagy for the Regeneration of the Aging Liver. Int J Mol Sci 2020; 21:ijms21103606. [PMID: 32443776 PMCID: PMC7279469 DOI: 10.3390/ijms21103606] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Age is one of the key risk factors to develop malignant diseases leading to a high incidence of hepatic tumors in the elderly population. The only curative treatment for hepatic tumors is surgical removal, which initiates liver regeneration. However, liver regeneration is impaired with aging, leading to an increased surgical risk for the elderly patient. Due to the increased risk, those patients are potentially excluded from curative surgery. Aging impairs autophagy via lipofuscin accumulation and inhibition of autophagosome formation. Autophagy is a recycling mechanism for eukaryotic cells to maintain homeostasis. Its principal function is to degrade endogenous bio-macromolecules for recycling cellular substances. A number of recent studies have shown that the reduced regenerative capacity of the aged remnant liver can be restored by promoting autophagy. Autophagy can be activated via multiple mTOR-dependent and mTOR-independent pathways. However, inducing autophagy through the mTOR-dependent pathway alone severely impairs liver regeneration. In contrast, recent observations suggest that inducing autophagy via mTOR-independent pathways might be promising in promoting liver regeneration. Conclusion: Activation of autophagy via an mTOR-independent autophagy inducer is a potential therapy for promoting liver regeneration, especially in the elderly patients at risk.
Collapse
Affiliation(s)
- Fengming Xu
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany; (F.X.); (C.H.); (H.-M.T.)
| | - Chuanfeng Hua
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany; (F.X.); (C.H.); (H.-M.T.)
| | - Hans-Michael Tautenhahn
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany; (F.X.); (C.H.); (H.-M.T.)
| | - Olaf Dirsch
- Institute of Pathology, Klinikum Chemnitz gGmbH, 09111 Chemnitz, Germany;
| | - Uta Dahmen
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany; (F.X.); (C.H.); (H.-M.T.)
- Correspondence: ; Tel.: +49-03641-9325350
| |
Collapse
|
13
|
Bangru S, Kalsotra A. Cellular and molecular basis of liver regeneration. Semin Cell Dev Biol 2020; 100:74-87. [PMID: 31980376 DOI: 10.1016/j.semcdb.2019.12.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Abstract
Recent advances in genetics and genomics have reinvigorated the field of liver regeneration. It is now possible to combine lineage-tracing with genome-wide studies to genetically mark individual liver cells and their progenies and detect precise changes in their genome, transcriptome, and proteome under normal versus regenerative settings. The recent use of single-cell RNA sequencing methodologies in model organisms has, in some ways, transformed our understanding of the cellular and molecular biology of liver regeneration. Here, we review the latest strides in our knowledge of general principles that coordinate regeneration of the liver and reflect on some conflicting evidence and controversies surrounding this topic. We consider the prominent mechanisms that stimulate homeostasis-related vis-à-vis injury-driven regenerative responses, highlight the likely cellular sources/depots that reconstitute the liver following various injuries and discuss the extrinsic and intrinsic signals that direct liver cells to proliferate, de-differentiate, or trans-differentiate while the tissue recovers from acute or chronic damage.
Collapse
Affiliation(s)
- Sushant Bangru
- Departments of Biochemistry and Pathology, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@ Illinois, University of Illinois, Urbana-Champaign, IL, USA
| | - Auinash Kalsotra
- Departments of Biochemistry and Pathology, University of Illinois, Urbana-Champaign, IL, USA; Cancer Center@ Illinois, University of Illinois, Urbana-Champaign, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, IL, USA.
| |
Collapse
|
14
|
Zhao X, Fang Y, Wang X, Yang Z, Li D, Tian M, Kang P. Knockdown of Ski decreases osteosarcoma cell proliferation and migration by suppressing the PI3K/Akt signaling pathway. Int J Oncol 2019; 56:206-218. [PMID: 31746363 PMCID: PMC6910224 DOI: 10.3892/ijo.2019.4914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
Ski, an evolutionary conserved protein, is involved in the development of a number of tumors, such as Barrett's esophagus, leukemia, colorectal cancer, gastric cancer, pancreatic cancer, hemangiomas and melanoma. However, studies on the functions of Ski in osteosarcoma (OS) are limited. In this study, firstly the differential expression of Ski in OS tissues and osteochondroma tissues was detected, and the expression of Ski in both human OS cell lines (MG63 and U2OS) and normal osteoblasts (hFoB1.19) was then detected. The results demonstrated that Ski expression was significantly upregulated in both human OS tissues and cell lines. The results led us to hypothesize that Ski may play an essential role in the pathological process of OS. Thus, Ski specific small interfere RNA (Ski-siRNA) was used. The results revealed that OS cell proliferation was markedly inhibited following the knockdown of Ski, which was identified by CCK8 assay, EdU staining and cell cycle analysis. In addition, OS cell migration was significantly suppressed following Ski knockdown, which was identified by wound healing assay. Moreover, the protein levels of p-PI3K and p-Akt in OS cells declined prominently following Ski knockdown. On the whole, the findings of this study revealed that Ski expression was significantly upregulated in OS tissue and OS cells. The knockdown of Ski decreased OS cell proliferation and migration, which was mediated by blocking the PI3K/Akt signaling pathway. Thus, Ski may act as a tumor promoter gene in tumorigenesis, and Ski may prove to be a potential therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuying Fang
- Weifang Maternal and Child Health Hospital, Weifang, Shandong 261000, P.R. China
| | - Xingwen Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Zhouyuan Yang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Donghai Li
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Meng Tian
- Neurosurgery Research Laboratory, West China Hospital, Sichuan Univerisity, Chengdu, Sichuan 610041, P.R. China
| | - Pengde Kang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
15
|
Zhao X, Wei Z, Li D, Yang Z, Tian M, Kang P. Glucocorticoid Enhanced the Expression of Ski in Osteonecrosis of Femoral Head: The Effect on Adipogenesis of Rabbit BMSCs. Calcif Tissue Int 2019; 105:506-517. [PMID: 31359074 DOI: 10.1007/s00223-019-00592-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/22/2019] [Indexed: 02/05/2023]
Abstract
Glucocorticoid (GC)-induced osteonecrosis has been considered as the most serious side effect in long-term or over-dose steroid therapy. The decreased bone mass and increased marrow fat tissue demonstrated that GC can destroy the normal differentiation of bone marrow mesenchymal stem cells (BMSCs), which accelerates adipogenesis but not osteogenesis. However, the underlying mechanisms are still unclear. Ski, an evolutionary conserved protein, is a multifunctional transcriptional regulator that involved in regulating signaling pathways associated with adipogenesis differentiation, but the concrete function remains unclear. In this work, we first established a methylprednisolone (MPS)-induced osteonecrosis of femoral head (ONFH) rabbit model, in which the expression of Ski, PPAR-γ, and FABP4 was up-regulated compared with control group, and then we induced the isolated BMSCs from rabbit with dexamethasone (Dex) in vitro and the results showed that the Ski expression was up-regulated by Dex in a dose- and time-dependent manner. Therefore, we demonstrated that the expression of Ski was up-regulated in glucocorticoid-related osteonecrosis disease in vivo and in vitro. Moreover, the adipogenesis differentiation capacity of BMSCs was enhanced after induced by Dex, which was identified by Oil Red O staining, and the up-regulated PPAR-γ and FABP4 expression. To further study the function of Ski in BMSC after induced by Dex, Ski specific small interfering RNA (Ski-siRNA) was used. Results showed that knockdown of Ski obviously decreased adipogenesis differentiation evident by Oil Red O staining, and the expression of PPAR-γ and FABP4 was down-regulated simultaneously. Collectively, our findings suggest that Ski increased significantly during glucocorticoid-induced adipogenic differentiation of BMSCs, and the expression level was consistent with adipogenic-related proteins including PPAR-γ and FABP4. Based on the above data, we believe that Ski might become a new molecule in the treatment of GC-induced ONFH and our study could provide a basis for further study on the detailed function of Ski in ONFH.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Zhun Wei
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Donghai Li
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Zhouyuan Yang
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China
| | - Meng Tian
- Neurosurgery Research Laboratory, West China Hospital, Sichuan Univerisity, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Pengde Kang
- Department of Orthopaedics Surgery, West China Hospital, Sichuan University, No. 37 Wainan Guoxue Road, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
16
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
17
|
Zhao X, Zhou K, Li Z, Nan W, Wang J, Xia Y, Zhang H. Knockdown of Ski decreased the reactive astrocytes proliferation in vitro induced by oxygen‐glucose deprivation/reoxygenation. J Cell Biochem 2018; 119:4548-4558. [DOI: 10.1002/jcb.26597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/07/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Xin Zhao
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Kai‐Sheng Zhou
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Zhong‐Hao Li
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Wei Nan
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Jing Wang
- Orthopaedics Key laboratory of Gansu ProvinceLanzhouPR China
| | - Ya‐Yi Xia
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
| | - Hai‐Hong Zhang
- The Second Clinical Medical College of Lanzhou UniversityLanzhouPR China
| |
Collapse
|
18
|
Abstract
Ski is an evolutionarily conserved protein and widely participates in the regulation of various pathological and physiological processes such as wound healing, liver regeneration, development of the embryonic nervous system, muscle differentiation, and progression of many kinds of tumors. However, the distribution and function of Ski in central nervous system lesion and disease remain unclear. In this study, we investigated the spatiotemporal expression of Ski in a spinal cord injury (SCI) model in adult rats. Western Blot analysis indicated that Ski was expressed in both normal and injured spinal cord, and showed a significant upregulation after SCI compared with the sham group. Double-labeled immunofluorescence staining showed that Ski was significantly expressed in astrocytes, but not in the neurons. Western Blot analyses of glial fibrillary acidic protein (GFAP) and BBB scores were carried out and correlation analysis showed a positive correlation between them. In addition, the relative expression level of Ski was also positively correlated with the relative expression level of GFAP. Moreover, the conspicuous co-expression band of Ski and GFAP at the lesion border was found in the results of immunofluorescence staining combined with the pattern of glial scar formation reflected by H&E staining; in addition, it was found that Ski was also highly associated with glial scar. On the basis of our data, we speculated that Ski might play an important role in the process of reactive astrogliosis after SCI and our study might provide a basis for further study on the detailed role of Ski in astrocytes.
Collapse
|
19
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Zhao X, Wang XW, Zhou KS, Nan W, Guo YQ, Kou JL, Wang J, Xia YY, Zhang HH. Expression of Ski and its role in astrocyte proliferation and migration. Neuroscience 2017; 362:1-12. [DOI: 10.1016/j.neuroscience.2017.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/13/2017] [Accepted: 08/14/2017] [Indexed: 10/19/2022]
|
21
|
Rao S, Zaidi S, Banerjee J, Jogunoori W, Sebastian R, Mishra B, Nguyen BN, Wu RC, White J, Deng C, Amdur R, Li S, Mishra L. Transforming growth factor-β in liver cancer stem cells and regeneration. Hepatol Commun 2017; 1:477-493. [PMID: 29404474 PMCID: PMC5678904 DOI: 10.1002/hep4.1062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/27/2017] [Accepted: 06/01/2017] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells have established mechanisms that contribute to tumor heterogeneity as well as resistance to therapy. Over 40% of hepatocellular carcinomas (HCCs) are considered to be clonal and arise from a stem-like/cancer stem cell. Moreover, HCC is the second leading cause of cancer death worldwide, and an improved understanding of cancer stem cells and targeting these in this cancer are urgently needed. Multiple studies have revealed etiological patterns and multiple genes/pathways signifying initiation and progression of HCC; however, unlike the transforming growth factor β (TGF-β) pathway, loss of p53 and/or activation of β-catenin do not spontaneously drive HCC in animal models. Despite many advances in cancer genetics that include identifying the dominant role of TGF-β signaling in gastrointestinal cancers, we have not reached an integrated view of genetic mutations, copy number changes, driver pathways, and animal models that support effective targeted therapies for these common and lethal cancers. Moreover, pathways involved in stem cell transformation into gastrointestinal cancers remain largely undefined. Identifying the key mechanisms and developing models that reflect the human disease can lead to effective new treatment strategies. In this review, we dissect the evidence obtained from mouse and human liver regeneration, and mouse genetics, to provide insight into the role of TGF-β in regulating the cancer stem cell niche. (Hepatology Communications 2017;1:477-493).
Collapse
Affiliation(s)
- Shuyun Rao
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Sobia Zaidi
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Jaideep Banerjee
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Wilma Jogunoori
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Raul Sebastian
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Bibhuti Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Bao-Ngoc Nguyen
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Ray-Chang Wu
- Department of Biochemistry and Molecular Medicine George Washington University Washington DC
| | - Jon White
- Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| | - Chuxia Deng
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Health Sciences University of Macau Taipa Macau China
| | - Richard Amdur
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC
| | - Shulin Li
- Department of Pediatrics The University of Texas MD Anderson Cancer Center Houston TX
| | - Lopa Mishra
- Center for Translational Medicine Department of Surgery, George Washington University Washington DC.,Institute for Clinical Research, Veterans Affairs Medical Center Washington DC
| |
Collapse
|
22
|
Liu M, Chen P. Proliferation‑inhibiting pathways in liver regeneration (Review). Mol Med Rep 2017; 16:23-35. [PMID: 28534998 DOI: 10.3892/mmr.2017.6613] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 03/13/2017] [Indexed: 12/14/2022] Open
Abstract
Liver regeneration, an orchestrated process, is the primary compensatory mechanism following liver injury caused by various factors. The process of liver regeneration consists of three stages: Initiation, proliferation and termination. Proliferation‑promoting factors, which stimulate the recovery of mitosis in quiescent hepatocytes, are essential in the initiation and proliferation steps of liver regeneration. Proliferation‑promoting factors act as the 'motor' of liver regeneration, whereas proliferation inhibitors arrest cell proliferation when the remnant liver reaches a suitable size. Certain proliferation inhibitors are also expressed and activated in the first two steps of liver regeneration. Anti‑proliferation factors, acting as a 'brake', control the speed of proliferation and determine the terminal point of liver regeneration. Furthermore, anti‑proliferation factors function as a 'steering‑wheel', ensuring that the regeneration process proceeds in the right direction by preventing proliferation in the wrong direction, as occurs in oncogenesis. Therefore, proliferation inhibitors to ensure safe and stable liver regeneration are as important as proliferation‑promoting factors. Cytokines, including transforming growth factor‑β and interleukin‑1, and tumor suppressor genes, including p53 and p21, are important members of the proliferation inhibitor family in liver regeneration. Certain anti‑proliferation factors are involved in the process of gene expression and protein modification. The suppression of liver regeneration led by metabolism, hormone activity and pathological performance have been reviewed previously. However, less is known regarding the proliferation inhibitors of liver regeneration and further investigations are required. Detailed information regarding the majority of known anti‑proliferation signaling pathways also remains fragmented. The present review aimed to understand the signalling pathways that inhbit proliferation in the process of liver regeneration.
Collapse
Affiliation(s)
- Menggang Liu
- Department of Hepatobiliary Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China
| | - Ping Chen
- Department of Hepatobiliary Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
23
|
de Jonge J, Olthoff KM. Liver regeneration. BLUMGART'S SURGERY OF THE LIVER, BILIARY TRACT AND PANCREAS, 2-VOLUME SET 2017:93-109.e7. [DOI: 10.1016/b978-0-323-34062-5.00006-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Tu W, Ye J, Wang ZJ. Embryonic liver fordin is involved in glucose glycolysis of hepatic stellate cell by regulating PI3K/Akt signaling. World J Gastroenterol 2016; 22:8519-8527. [PMID: 27784964 PMCID: PMC5064033 DOI: 10.3748/wjg.v22.i38.8519] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/27/2016] [Accepted: 08/19/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the role of embryonic liver fordin (ELF) in liver fibrosis by regulating hepatic stellate cells (HSCs) glucose glycolysis.
METHODS The expression of ELF and the glucose glycolysis-related proteins were evaluated in activated HSCs. siRNA was used to silence ELF expression in activated HSCs in vitro and the subsequent changes in PI3K/Akt signaling and glucose glycolysis-related proteins were observed.
RESULTS The expression of ELF increased remarkably in HSCs of the fibrosis mouse model and HSCs that were cultured for 3 wk in vitro. Glucose glycolysis-related proteins showed an obvious increase in the activated HSCs, such as phosphofructokinase, platelet and glucose transporter 1. ELF-siRNA, which perfectly silenced the expression of ELF in activated HSCs, led to the induction of glucose glycolysis-related proteins and extracellular matrix (ECM) components. Moreover, pAkt, which is an important downstream factor in PI3K/Akt signaling, showed a significant change in response to the ELF silencing. The expression of glucose glycolysis-related proteins and ECM components decreased remarkably when the PI3K/Akt signaling was blocked by Ly294002 in the activated HSCs.
CONCLUSION ELF is involved in HSC glucose glycolysis by regulating PI3K/Akt signaling.
Collapse
|
25
|
Caligaris C, Vázquez-Victorio G, Sosa-Garrocho M, Ríos-López DG, Marín-Hernández A, Macías-Silva M. Actin-cytoskeleton polymerization differentially controls the stability of Ski and SnoN co-repressors in normal but not in transformed hepatocytes. Biochim Biophys Acta Gen Subj 2015; 1850:1832-41. [PMID: 26002202 DOI: 10.1016/j.bbagen.2015.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ski and SnoN proteins function as transcriptional co-repressors in the TGF-β pathway. They regulate cell proliferation and differentiation, and their aberrant expression results in altered TGF-β signalling, malignant transformation, and alterations in cell proliferation. METHODS We carried out a comparative characterization of the endogenous Ski and SnoN protein regulation by TGF-β, cell adhesion disruption and actin-cytoskeleton rearrangements between normal and transformed hepatocytes; we also analyzed Ski and SnoN protein stability, subcellular localization, and how their protein levels impact the TGF-β/Smad-driven gene transcription. RESULTS Ski and SnoN protein levels are lower in normal hepatocytes than in hepatoma cells. They exhibit a very short half-life and a nuclear/cytoplasmic distribution in normal hepatocytes opposed to a high stability and restricted nuclear localization in hepatoma cells. Interestingly, while normal cells exhibit a transient TGF-β-induced gene expression, the hepatoma cells are characterized by a strong and sustained TGF-β-induced gene expression. A novel finding is that Ski and SnoN stability is differentially regulated by cell adhesion and cytoskeleton rearrangements in the normal hepatocytes. The inhibition of protein turnover down-regulated both Ski and SnoN co-repressors impacting the kinetic of expression of TGF-β-target genes. CONCLUSION Normal regulatory mechanisms controlling Ski and SnoN stability, subcellular localization and expression are altered in hepatocarcinoma cells. GENERAL SIGNIFICANCE This work provides evidence that Ski and SnoN protein regulation is far more complex in normal than in transformed cells, since many of the normal regulatory mechanisms are lost in transformed cells.
Collapse
Affiliation(s)
- Cassandre Caligaris
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Genaro Vázquez-Victorio
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Marcela Sosa-Garrocho
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Diana G Ríos-López
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Alvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México D.F., 14080, México
| | - Marina Macías-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México.
| |
Collapse
|
26
|
Vázquez-Victorio G, Caligaris C, Del Valle-Espinosa E, Sosa-Garrocho M, González-Arenas NR, Reyes-Cruz G, Briones-Orta MA, Macías-Silva M. Novel regulation of Ski protein stability and endosomal sorting by actin cytoskeleton dynamics in hepatocytes. J Biol Chem 2015; 290:4487-99. [PMID: 25561741 DOI: 10.1074/jbc.m114.579532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TGF-β-induced antimitotic signals are highly regulated during cell proliferation under normal and pathological conditions, such as liver regeneration and cancer. Up-regulation of the transcriptional cofactors Ski and SnoN during liver regeneration may favor hepatocyte proliferation by inhibiting TGF-β signals. In this study, we found a novel mechanism that regulates Ski protein stability through TGF-β and G protein-coupled receptor (GPCR) signaling. Ski protein is distributed between the nucleus and cytoplasm of normal hepatocytes, and the molecular mechanisms controlling Ski protein stability involve the participation of actin cytoskeleton dynamics. Cytoplasmic Ski is partially associated with actin and localized in cholesterol-rich vesicles. Ski protein stability is decreased by TGF-β/Smads, GPCR/Rho signals, and actin polymerization, whereas GPCR/cAMP signals and actin depolymerization promote Ski protein stability. In conclusion, TGF-β and GPCR signals differentially regulate Ski protein stability and sorting in hepatocytes, and this cross-talk may occur during liver regeneration.
Collapse
Affiliation(s)
- Genaro Vázquez-Victorio
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Cassandre Caligaris
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Eugenio Del Valle-Espinosa
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Marcela Sosa-Garrocho
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Nelly R González-Arenas
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Guadalupe Reyes-Cruz
- the Departamento de Biología Celular, CINVESTAV-IPN, México, D. F. 07000, México
| | - Marco A Briones-Orta
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Marina Macías-Silva
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| |
Collapse
|
27
|
Arno AI, Gauglitz GG, Barret JP, Jeschke MG. New molecular medicine-based scar management strategies. Burns 2014; 40:539-51. [PMID: 24438742 DOI: 10.1016/j.burns.2013.11.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/21/2013] [Accepted: 11/18/2013] [Indexed: 02/06/2023]
Abstract
Keloids and hypertrophic scars are prevalent disabling conditions with still suboptimal treatments. Basic science and molecular-based medicine research have contributed to unravel new bench-to-bedside scar therapies and to dissect the complex signalling pathways involved. Peptides such as the transforming growth factor beta (TGF-β) superfamily, with Smads, Ski, SnoN, Fussels, endoglin, DS-Sily, Cav-1p, AZX100, thymosin-β4 and other related molecules may emerge as targets to prevent and treat keloids and hypertrophic scars. The aim of this review is to describe the basic complexity of these new molecular scar management strategies and point out new fibrosis research lines.
Collapse
Affiliation(s)
- Anna I Arno
- Ross Tilley Burn Centre and Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Gerd G Gauglitz
- Department of Dermatology and Allergology, Ludwig Maximilians University, Munich, Germany
| | - Juan P Barret
- Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Marc G Jeschke
- Ross Tilley Burn Centre and Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
28
|
A20-An Omnipotent Protein in the Liver: Prometheus Myth Resolved? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 809:117-39. [DOI: 10.1007/978-1-4939-0398-6_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Zhang R, Sun J, Zhang Y, Cheng S, Zhang X. Signal transduction disturbance related to hepatocarcinogenesis in mouse by prolonged exposure to Nanjing drinking water. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2013; 20:6468-6481. [PMID: 23591932 DOI: 10.1007/s11356-013-1695-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Accepted: 03/28/2013] [Indexed: 06/02/2023]
Abstract
Toxicogenomic approaches were used to investigate the potential hepatocarcinogenic effects on mice by oral exposure to Nanjing drinking water (NJDW). Changes in the hepatic transcriptome of 3 weeks male mice (Mus musculus) were monitored and dissected after oral exposure to NJDW for 90 days. No preneoplastic and neoplastic lesions were observed in the hepatic tissue by the end of NJDW exposure. However, total of 746 genes were changed transcriptionally. Thirty-one percent of differentially expressed genes (DEGs) were associated with the functional categories of cell cycle regulation, adhesion, growth, apoptosis, and signal transduction, which are closely implicated in tumorigenesis and progression. Interrogation of Kyoto Encyclopedia of Genes and Genomes revealed that 43 DEGs were mapped to several crucial signaling pathways implicated in the pathogenesis of hepatocellular carcinoma (HCC). In signal transduction network constructed via Genes2Networks software, Egfr, Akt1, Atf2, Ctnnb1, Hras, Mapk1, Smad2, and Ccnd1 were hubs. Direct gene-disease relationships obtained from Comparative Toxicogenomics Database and scientific literatures revealed that the hubs have direct mechanism or biomarker relationships with hepatocellular preneoplastic lesions or hepatocarcinogenesis. Therefore, prolonged intake of NJDW without employing any indoor water treatment strategy might predispose mouse to HCC. Furthermore, Egfr, Akt1, Ctnnb1, Hras, Mapk1, Smad2, and Ccnd1 were identified as promising biomarkers of the potential combined hepatocarcinogenicity.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Road, Nanjing, 210046, People's Republic of China
| | | | | | | | | |
Collapse
|
30
|
Hernández-Damián J, Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Vázquez-Macías A, Caligaris C, Sosa-Garrocho M, Flores-Pérez B, Romero-Avila M, Macías-Silva M. Downregulation of SnoN oncoprotein induced by antibiotics anisomycin and puromycin positively regulates transforming growth factor-β signals. Biochim Biophys Acta Gen Subj 2013; 1830:5049-58. [PMID: 23872350 DOI: 10.1016/j.bbagen.2013.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/26/2013] [Accepted: 07/09/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND SnoN and Ski proteins function as Smad transcriptional corepressors and are implicated in the regulation of diverse cellular processes such as proliferation, differentiation and transformation. Transforming growth factor-β (TGF-β) signaling causes SnoN and Ski protein degradation via proteasome with the participation of phosphorylated R-Smad proteins. Intriguingly, the antibiotics anisomycin (ANS) and puromycin (PURO) are also able to downregulate Ski and SnoN proteins via proteasome. METHODS We explored the effects of ANS and PURO on SnoN protein downregulation when the activity of TGF-β signaling was inhibited by using different pharmacological and non-pharmacological approaches, either by using specific TβRI inhibitors, overexpressing the inhibitory Smad7 protein, or knocking-down TβRI receptor or Smad2 by specific shRNAs. The outcome of SnoN and Ski downregulation induced by ANS or PURO on TGF-β signaling was also studied. RESULTS SnoN protein downregulation induced by ANS and PURO did not involve the induction of R-Smad phosphorylation but it was abrogated after TGF-β signaling inhibition; this effect occurred in a cell type-specific manner and independently of protein synthesis inhibition or any other ribotoxic effect. Intriguingly, antibiotics seem to require components of the TGF-β/Smad pathway to downregulate SnoN. In addition, SnoN protein downregulation induced by antibiotics favored gene transcription induced by TGF-β signaling. CONCLUSIONS ANS and PURO require TGF-β/Smad pathway to induce SnoN and Ski protein downregulation independently of inducing R-Smad2 phosphorylation, which facilitates TGF-β signaling. GENERAL SIGNIFICANCE Antibiotic analogs lacking ribotoxic effects are useful as pharmacological tools to study TGF-β signaling by controlling Ski and SnoN protein levels.
Collapse
Affiliation(s)
- Jacqueline Hernández-Damián
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., 04510 Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nygård IE, Mortensen KE, Hedegaard J, Conley LN, Kalstad T, Bendixen C, Revhaug A. The genetic regulation of the terminating phase of liver regeneration. COMPARATIVE HEPATOLOGY 2012; 11:3. [PMID: 23164283 PMCID: PMC3558440 DOI: 10.1186/1476-5926-11-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 11/08/2012] [Indexed: 02/06/2023]
Abstract
UNLABELLED BACKGROUND After partial hepatectomy (PHx), the liver regeneration process terminates when the normal liver-mass/body-weight ratio of 2.5% has been re-established. To investigate the genetic regulation of the terminating phase of liver regeneration, we performed a 60% PHx in a porcine model. Liver biopsies were taken at the time of resection, after three weeks and upon termination the sixth week. Gene expression profiles were obtained using porcine oligonucleotide microarrays. Our study reveals the interactions between genes regulating the cell cycle, apoptosis and angiogenesis, and the role of Transforming Growth Factor-β (TGF-β) signalling towards the end of liver regeneration. RESULTS Microarray analysis revealed a dominance of genes regulating apoptosis towards the end of regeneration. Caspase Recruitment Domain-Containing Protein 11 (CARD11) was up-regulated six weeks after PHx, suggesting the involvement of the caspase system at this time. Zinc Finger Protein (ZNF490) gene, with a potential negative effect on cell cycle progression, was only up-regulated at three and six weeks after PHx indicating a central role at this time. TGF-β regulation was not found to be significantly affected in the terminating phase of liver regeneration. Vasohibin 2 (VASH2) was down-regulated towards the end of regeneration, and may indicate a role in preventing a continued vascularization process. CONCLUSIONS CARD11, ZNF490 and VASH2 are differentially expressed in the termination phase of liver regeneration. The lack of TGF-β up-regulation suggests that signalling by TGF-β is not required for termination of liver regeneration.
Collapse
Affiliation(s)
- Ingvild E Nygård
- Department of Digestive Surgery, University Hospital of Northern-Norway, Tromsø 9038, Norway.
| | | | | | | | | | | | | |
Collapse
|
32
|
Parlakgumus A, Colakoglu T, Kayaselcuk F, Colakoglu S, Ezer A, Calıskan K, Karakaya J, Yildirim S. Two drugs with paradoxical effects on liver regeneration through antiangiogenesis and antifibrosis: Losartan and Spironolactone: a pharmacologic dilemma on hepatocyte proliferation. J Surg Res 2012; 179:60-5. [PMID: 22989552 DOI: 10.1016/j.jss.2012.08.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 07/06/2012] [Accepted: 08/22/2012] [Indexed: 01/12/2023]
Abstract
BACKGROUND There is a strong relationship between liver regeneration and angiogenesis and fibrosis. It is known that Spironolactone, an aldosterone antagonist, acting on rennin-aldosterone axis, and Losartan, an angiotensin II type I antagonist, have both antifibrotic and antiangiogenic effects. Theoretically, the end result of these mechanisms with contradictory influences on liver regeneration is not known well. In this study, we aimed to reveal the effects on liver regeneration of administration of Spironolactone and Losartan, having contradicting effects on regeneration through antiangiogenesis and antifibrosis. MATERIALS AND METHODS A total of 72 Wistar albino rats were divided into control, Spironolactone, and Losartan groups and subdivided to conduct examinations on days 1, 3, 5, and 7. The specimens were treated with proliferating cell nuclear antigen to evaluate the characteristics of liver regeneration; with phosphorylated Smad2 (phospho-Smad2), serum transforming growth factor beta (TGF-B) 1, and tissue TGF-B1 to evaluate the termination of regeneration and with vascular endothelial growth factor receptor 2, Flk-1/KDR, to evaluate angiogenesis. RESULTS The proliferating cell nuclear antigen-labeling index was found to be significantly higher in Spironolactone and Losartan groups than in the control group on days 1, 3, and 5 (P = 0.031, 0.0023, and 0.032, respectively). Vascular endothelial growth factor receptor 2, Flk-1/KDR, expression was significantly lower in Spironolactone and Losartan groups than in the control group on days 3, 5, and 7 (P = 0.032, 0.0024, and 0.007, respectively). Phospho-Smad2 was significantly lower on days 1, 3, and 5 in Spironolactone and Losartan groups than in the control group (P = 0.011, 0.0020, and 0.05, respectively). Tissue TGF-B1 levels were significantly lower in Spironolactone and Losartan groups than in the control group only on day 3 (P = 0039). Serum TGF-B1 levels in Losartan groups were significantly different from those of control and Spironolactone groups only on day 1 (P < 0.05). CONCLUSIONS Liver regeneration, expected to decrease on day 3, was prolonged and increased even on day 5 despite antiangiogenic effects of Losartan and Spironolactone, which in fact inhibit fibrosis through phospho-Smad2 and increase regeneration. In addition, serum and tissue TGF-B1 levels are not sensitive enough to show active TGF-B1 for the evaluation of regeneration.
Collapse
Affiliation(s)
- Alper Parlakgumus
- Department of General Surgery, Adana Teaching and Research Center, Baskent University School of Medicine, Ankara, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Tecalco-Cruz AC, Sosa-Garrocho M, Vázquez-Victorio G, Ortiz-García L, Domínguez-Hüttinger E, Macías-Silva M. Transforming growth factor-β/SMAD Target gene SKIL is negatively regulated by the transcriptional cofactor complex SNON-SMAD4. J Biol Chem 2012; 287:26764-76. [PMID: 22674574 PMCID: PMC3411014 DOI: 10.1074/jbc.m112.386599] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Indexed: 12/23/2022] Open
Abstract
The human SKI-like (SKIL) gene encodes the SMAD transcriptional corepressor SNON that antagonizes TGF-β signaling. SNON protein levels are tightly regulated by the TGF-β pathway: whereas a short stimulation with TGF-β decreases SNON levels by its degradation via the proteasome, longer TGF-β treatment increases SNON levels by inducing SKIL gene expression. Here, we investigated the molecular mechanisms involved in the self-regulation of SKIL gene expression by SNON. Bioinformatics analysis showed that the human SKIL gene proximal promoter contains a TGF-β response element (TRE) bearing four groups of SMAD-binding elements that are also conserved in mouse. Two regions of 408 and 648 bp of the human SKIL gene (∼2.4 kb upstream of the ATG initiation codon) containing the core promoter, transcription start site, and the TRE were cloned for functional analysis. Binding of SMAD and SNON proteins to the TRE region of the SKIL gene promoter after TGF-β treatment was demonstrated by ChIP and sequential ChIP assays. Interestingly, the SNON-SMAD4 complex negatively regulated basal SKIL gene expression through binding the promoter and recruiting histone deacetylases. In response to TGF-β signal, SNON is removed from the SKIL gene promoter, and then the activated SMAD complexes bind the promoter to induce SKIL gene expression. Subsequently, the up-regulated SNON protein in complex with SMAD4 represses its own expression as part of the negative feedback loop regulating the TGF-β pathway. Accordingly, when the SNON-SMAD4 complex is absent as in some cancer cells lacking SMAD4 the regulation of some TGF-β target genes is modified.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Marcela Sosa-Garrocho
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Genaro Vázquez-Victorio
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Layla Ortiz-García
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Elisa Domínguez-Hüttinger
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| | - Marina Macías-Silva
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, Distrito Federal 04510, México
| |
Collapse
|
34
|
Wang Z, Song Y, Tu W, He X, Lin J, Liu F. β-2 spectrin is involved in hepatocyte proliferation through the interaction of TGFβ/Smad and PI3K/AKT signalling. Liver Int 2012; 32:1103-1111. [PMID: 22541060 DOI: 10.1111/j.1478-3231.2012.02812.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 03/22/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND Transforming growth factor (TGF) β signalling pathway plays a crucial role in liver regeneration following partial hepatectomy in mice. Evidence demonstrated that β-2 Spectrin is involved in TGFβ/Smad signalling pathway as a Smad3/4 adaptor protein. AIM The aim of this study was to explore the role of β-2 Spectrin in hepatocyte proliferation. METHODS β-2 Spectrin expression was evaluated in mice receiving partial hepatectomy. The effect of siRNA against β-2 Spectrin on hepatocyte proliferation was determined. The interaction between TGFβ/Smad and PI3K/Akt signalling was investigated. RESULTS Hepatic β-2 Spectrin decreased dramatically 2 days after 70% hepatectomy in mice. In AML-12 cells, hepatocyte proliferation was inhibited after the stimulation of TGF β1 and a reduction in β-2 Spectrin mediated by siRNA resulted in increase in proliferative response. Confocal results revealed that β-2 Spectrin represented a key regulator in TGFβ/Smad signalling through controlling Smad3/4 subcellular localization. Moreover, Alternation of Akt phosphorylation led to the change in subcellular localization of Smad2, 3, 4 and β-2 Spectrin, A reduction in Smad2, 3 and 4 mediated by siRNA resulted in the induction of pAkt expression. CONCLUSIONS These findings reveal that β-2 Spectrin plays a crucial role in hepatocyte proliferation, which contributes to liver regeneration following hepatectomy in mice. In addition, PI3K/Akt is involved in TGFβ/Smad signalling pathway through the interaction with Smad proteins and β-2 Spectrin.
Collapse
Affiliation(s)
- Zhijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
The liver is the body's most important detoxification organ and has an extreme ability to regenerate. The regeneration process can be divided into three stages: initiation, proliferation and termination. Most of previous studies focus on the initial stage and proliferative stage, while the mechanism for the proper termination of liver regeneration is still poorly understood. The termination stage involves a variety of cytokines and growth factors, which mainly function to inhibit mitogen-mediated liver cell growth-promoting effect and promote the apoptosis of excessively proliferating liver cells. In this paper we will discuss the major factors involved in the termination of liver regeneration.
Collapse
|
36
|
Zhu Q, Luo K. SnoN in regulation of embryonic development and tissue morphogenesis. FEBS Lett 2012; 586:1971-6. [PMID: 22710172 DOI: 10.1016/j.febslet.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/03/2012] [Accepted: 03/05/2012] [Indexed: 01/15/2023]
Abstract
SnoN (Ski-novel protein) plays an important role in embryonic development, tumorigenesis and aging. Past studies largely focused on its roles in tumorigenesis. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed that SnoN interacts with multiple signaling molecules at different cellular levels to modulate the activities of several signaling pathways in a tissue context and developmental stage dependent manner. These studies suggest that SnoN may have broad functions in the embryonic development and tissue morphogenesis.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
37
|
Crawford JM, Burt AD. Anatomy, pathophysiology and basic mechanisms of disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2012:1-77. [DOI: 10.1016/b978-0-7020-3398-8.00001-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
38
|
Jonge JD, Olthoff KM. Liver regeneration. BLUMGART'S SURGERY OF THE LIVER, PANCREAS AND BILIARY TRACT 2012:87-101.e6. [DOI: 10.1016/b978-1-4377-1454-8.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
39
|
Pagano MA, Tibaldi E, Gringeri E, Brunati AM. Tyrosine phosphorylation and liver regeneration: A glance at intracellular transducers. IUBMB Life 2011; 64:27-35. [DOI: 10.1002/iub.576] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 08/15/2011] [Indexed: 12/30/2022]
|
40
|
Delgado-Coello B, Briones-Orta MA, Macías-Silva M, Mas-Oliva J. Cholesterol: recapitulation of its active role during liver regeneration. Liver Int 2011; 31:1271-84. [PMID: 21745289 DOI: 10.1111/j.1478-3231.2011.02542.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver regeneration is a compensatory hyperplasia produced by several stimuli that promotes proliferation in order to provide recovery of the liver mass and architecture. This process involves complex signalling cascades that receive feedback from autocrine and paracrine pathways, recognized by parenchymal as well as non-parenchymal cells. Nowadays the dynamic role of lipids in biological processes is widely recognized; however, a systematic analysis of their importance during liver regeneration is still missing. Therefore, in this review we address the role of lipids including the bioactive ones such as sphingolipids, but with special emphasis on cholesterol. Cholesterol is not only considered as a structural component but also as a relevant lipid involved in the control of the intermediate metabolism of different liver cell types such as hepatocytes, hepatic stellate cells and Kupffer cells. Cholesterol plays a significant role at the level of specific membrane domains, as well as modulating the expression of sterol-dependent proteins. Moreover, several enzymes related to the catabolism of cholesterol and whose activity is down regulated are related to the protection of liver tissue from toxicity during the process of regeneration. This review puts in perspective the necessity to study and understand the basic mechanisms involving lipids during the process of liver regeneration. On the other hand, the knowledge acquired in this area in the past years, can be considered invaluable in order to provide further insights into processes such as general organogenesis and several liver-related pathologies, including steatosis and fibrosis.
Collapse
Affiliation(s)
- Blanca Delgado-Coello
- Departamento de Bioquímica y Biología Estructural, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, DF Mexico
| | | | | | | |
Collapse
|
41
|
Kim RD, Kim JS, Watanabe G, Mohuczy D, Behrns KE. Liver regeneration and the atrophy-hypertrophy complex. Semin Intervent Radiol 2011; 25:92-103. [PMID: 21326550 DOI: 10.1055/s-2008-1076679] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The atrophy-hypertrophy complex (AHC) refers to the controlled restoration of liver parenchyma following hepatocyte loss. Different types of injury (e.g., toxins, ischemia/reperfusion, biliary obstruction, and resection) elicit the same hypertrophic response in the remnant liver. The AHC involves complex anatomical, histological, cellular, and molecular processes. The signals responsible for these processes are both intrinsic and extrinsic to the liver and involve both physical and molecular events. In patients in whom resection of large liver malignancies would result in an inadequate functional liver remnant, preoperative portal vein embolization may increase the remnant liver sufficiently to permit aggressive resections. Through continued basic science research, the cellular mechanisms of the AHC may be maximized to permit curative resections in patients with potentially prohibitive liver function.
Collapse
Affiliation(s)
- Robin D Kim
- Department of Surgery, Division of General and GI Surgery, University of Florida, Gainesville, Florida
| | | | | | | | | |
Collapse
|
42
|
Zhang R, Cheng S, Li A, Sun J, Zhang Y, Zhang X. Genome-wide screening of indicator genes for assessing the potential carcinogenic risk of Nanjing city drinking water. ECOTOXICOLOGY (LONDON, ENGLAND) 2011; 20:1033-1040. [PMID: 21424719 DOI: 10.1007/s10646-011-0647-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/13/2011] [Indexed: 05/30/2023]
Abstract
Effects of all pollutants existing in the Nanjing city drinking water (DWNC) on mouse gene transcription levels were measured to assess the DWNC carcinogenic risks and to identify candidate indicator genes for assessing and early warning the cancer risks. Transcriptional expression levels of 14,000 hepatic genes for the treatment group mice (Mus musculus, ICR) fed with DWNC for 90 days were detected using the GeneChip(®) Mouse Genome 430A 2.0 array. The analysis indicated that the transcriptional levels of 294 genes were up-regulated and 542 ones were down-regulated. Of these genes, 12 ones identified to be involved in at least five different types of cancers were further analyzed. An interrogation by Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that three (including ITGAV, CCND1 and SMAD2) of the 12 genes were mapped to pathway in cancer. Gene Ontology (GO) function annotation also showed that they were associated with the functional categories of cell cycle regulation, adhesion, apoptosis, signal transduction and so on which are closely implicated in tumorigenesis and progression. The correlations between the aberrant expressions of them and the genesis and progression of cancers have been further documented by a number of scientific researches. These results might demonstrate that the potential toxicity and carcinogenic risks were associated with DWNC. Moreover, ITGAV, CCND1 and SMAD2 were identified as the most likely candidate indicator genes for the assessment of the combined carcinogenic risk of all pollutants existing in DWNC.
Collapse
Affiliation(s)
- Rui Zhang
- State Key Laboratory of Pollution Control & Resource Reuse, School of the Environment, Nanjing University, 163 Xianlin Road, Nanjing, 210046, People's Republic of China
| | | | | | | | | | | |
Collapse
|
43
|
Bonnon C, Atanasoski S. c-Ski in health and disease. Cell Tissue Res 2011; 347:51-64. [DOI: 10.1007/s00441-011-1180-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/15/2011] [Indexed: 01/28/2023]
|
44
|
Thenappan A, Shukla V, Khalek FJA, Li Y, Shetty K, Liu P, Li L, Johnson RL, Johnson L, Mishra L. Loss of transforming growth factor β adaptor protein β-2 spectrin leads to delayed liver regeneration in mice. Hepatology 2011; 53:1641-50. [PMID: 21520177 PMCID: PMC3162320 DOI: 10.1002/hep.24111] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
UNLABELLED Liver regeneration, following partial hepatectomy (PHx), occurs through precisely controlled and synchronized cell proliferation, in which quiescent hepatocytes undergo one to two rounds of replication, with restoration of liver mass and function. We previously demonstrated that loss of the Smad3/4 adaptor protein β-2 spectrin (β2SP) is associated with faster entry into S phase, and hepatocellular cancer formation. These observations led us to further pursue the role of β2SP in cell cycle progression in vivo. Liver regeneration studies with PHx in β2SP(+/-) mice reveal a surprising and significant decrease in liver/body weight ratio at 48 hours after PHx in β2SP(+/-) mice in comparison to wildtype mice. At 48 hours after PHx we also observe decreased levels of cyclin E (2.4-fold, P < 0.05), Cdk1 (7.2-fold, P < 0.05), cyclin A, pRb (Ser249/Thr252), proliferative cell nuclear antigen (PCNA), cyclin D1 with elevated levels of pCdk1 (Thr14) (3.6-fold, P < 0.05). Strikingly, at 24 hours elevated levels of p53 (4-fold, P < 0.05), phospho-p53 (ser15 and ser20), and p21 (200-fold, P < 0.05) persisting to 48 hours after PHx further correlated with raised expression of the DNA damage markers pChk2 (Thr68) and γH2AX (S139). However, compromised cell cycle progression with loss of β2SP is not rescued by inhibiting p53 function, and that G(2) /M phase arrest observed is independent and upstream of p53. CONCLUSION β2SP deficiency results in dysfunctional hepatocyte cell cycle progression and delayed liver regeneration at 48 hours after PHx, which is p53-independent. β2SP loss may increase susceptibility to DNA damage, impair cell cycle progression, and ultimately lead to hepatocellular cancer.
Collapse
Affiliation(s)
- Arun Thenappan
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Vivek Shukla
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Feras J Abdul Khalek
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Ying Li
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirti Shetty
- Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington, DC
| | - Pu Liu
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lu Li
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Randy L Johnson
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lynt Johnson
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington, DC
| | - Lopa Mishra
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas,Correspondence to: Lopa Mishra, Lopa Mishra, MD, Del & Dennis McCarthy Distinguished Professor and Chair, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1466, Houston, TX, 77030, Tel: 202-687-5707, Fax: 202-687-0992,
| |
Collapse
|
45
|
Nechemia-Arbely Y, Shriki A, Denz U, Drucker C, Scheller J, Raub J, Pappo O, Rose-John S, Galun E, Axelrod JH. Early hepatocyte DNA synthetic response posthepatectomy is modulated by IL-6 trans-signaling and PI3K/AKT activation. J Hepatol 2011; 54:922-9. [PMID: 21145830 DOI: 10.1016/j.jhep.2010.08.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 07/05/2010] [Accepted: 08/03/2010] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Interleukin-6 (IL-6) is a crucial factor in liver regeneration following partial hepatectomy (PH); however, the role of IL-6 and IL-6 trans-signaling in particular, in hepatocyte mitosis remains controversial. IL-6 trans-signaling relies upon the release of the soluble IL-6R (sIL-6R), which binds IL-6 to form an agonistic IL-6/sIL-6R complex. Herein we have examined the hypothesis that IL-6 trans-signaling plays a crucial and distinct role in liver regeneration following PH. METHODS The specific IL-6/sIL-6R antagonist, sgp130Fc, was expressed in mice and analyzed for its effect on hepatocyte mitosis following PH. Alternatively, we examined the effect of the IL-6/sIL-6R super-agonist, Hyper-IL-6, or IL-6 expressed either alone or in combination with hepatocyte growth factor (HGF) on hepatocyte mitosis in the absence of PH. RESULTS Following PH, the dramatic rise of circulating IL-6 levels is accompanied by a concurrent ∼2-fold increase in circulating sIL-6R levels. Ectopic expression of sgp130Fc reduced hepatocyte mitosis by about 40% at early times following PH, while substantially reducing AKT, but not STAT3, activation. But, ectopic Hyper-IL-6 expression in mice without PH was not mitogenic to hepatocytes in vivo. Rather, Hyper-IL-6, but not IL-6, markedly increased HGF-induced hepatocyte mitosis. This cooperative effect correlated with greater resistance of HIL-6 than IL-6 to HGF-mediated reduction of AKT activation, rather than changes in STAT3 or MAPK signaling, and was completely blocked by PI3K inhibition. CONCLUSIONS Following PH, IL-6/sIL-6R cooperates with growth factors, through a PI3K/AKT-dependent mechanism to promote entry of hepatocytes into the cell cycle.
Collapse
Affiliation(s)
- Yael Nechemia-Arbely
- The Goldyne Savad Institute of Gene Therapy, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Revuelta-Cervantes J, Mayoral R, Miranda S, González-Rodríguez A, Fernández M, Martín-Sanz P, Valverde AM. Protein Tyrosine Phosphatase 1B (PTP1B) deficiency accelerates hepatic regeneration in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1591-604. [PMID: 21406170 DOI: 10.1016/j.ajpath.2010.12.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 12/09/2010] [Accepted: 12/17/2010] [Indexed: 01/27/2023]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a key regulator of metabolism and cell growth by its ability to dephosphorylate tyrosine kinase receptors and modulate the intensity of their signaling cascades. Because liver regeneration involves tyrosine phosphorylation-mediated signaling, we investigated the role of PTP1B in this process by performing partial hepatectomy in wild-type (PTP1B(+/+)) and PTP1B-deficient (PTP1B(-/-)) mice. The expression of PCNA and cyclins D1 and E (cell proliferation markers) was enhanced in PTP1B(-/-) regenerating livers, in parallel with 5'-bromo-2'-deoxyuridine incorporation. Phosphorylation of JNK1/2 and STAT3, early triggers of hepatic regeneration in response to TNF-α and IL-6, was accelerated in PTP1B(-/-) mice compared with PTP1B(+/+) mice. These phosphorylations were increased in PTP1B(-/-) hepatocytes or by silencing PTP1B in wild-type cells and decreased further after the addition of recombinant PTP1B. Enhanced EGF- and HGF receptor-mediated signaling was observed in regenerating livers lacking PTP1B and in EGF- or HGF-stimulated PTP1B(-/-) hepatocytes. Moreover, PTP1B(-/-) mice displayed a more rapid increase in intrahepatic lipid accumulation than PTP1B(+/+) control mice. Late responses to partial hepatectomy revealed additional divergences because stress-mediated signaling was attenuated at 24 to 96 hours in PTP1B(-/-) mice compared with PTP1B(+/+) mice. Finally, PTP1B deficiency also improves hepatic regeneration in mice fed a high-fat diet. These results suggest that pharmacological inhibition of PTP1B would improve liver regeneration in patients with acute or chronic liver injury.
Collapse
Affiliation(s)
- Jesús Revuelta-Cervantes
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
47
|
Li P, Liu P, Xiong RP, Chen XY, Zhao Y, Lu WP, Liu X, Ning YL, Yang N, Zhou YG. Ski, a modulator of wound healing and scar formation in the rat skin and rabbit ear. J Pathol 2011; 223:659-71. [DOI: 10.1002/path.2831] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 11/08/2010] [Accepted: 11/24/2010] [Indexed: 02/01/2023]
|
48
|
Böhm F, Köhler UA, Speicher T, Werner S. Regulation of liver regeneration by growth factors and cytokines. EMBO Mol Med 2010; 2:294-305. [PMID: 20652897 PMCID: PMC3377328 DOI: 10.1002/emmm.201000085] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The capability of the liver to fully regenerate after injury is a unique phenomenon essential for the maintenance of its important functions in the control of metabolism and xenobiotic detoxification. The regeneration process is histologically well described, but the genes that orchestrate liver regeneration have been only partially characterized. Of particular interest are cytokines and growth factors, which control different phases of liver regeneration. Historically, their potential functions in this process were addressed by analyzing their expression in the regenerating liver of rodents. Some of the predicted roles were confirmed using functional studies, including systemic delivery of recombinant growth factors, neutralizing antibodies or siRNAs prior to liver injury or during liver regeneration. In particular, the availability of genetically modified mice and their use in liver regeneration studies has unraveled novel and often unexpected functions of growth factors, cytokines and their downstream signalling targets in liver regeneration. This review summarizes the results obtained by functional studies that have addressed the roles and mechanisms of action of growth factors and cytokines in liver regeneration after acute injury to this organ.
Collapse
Affiliation(s)
- Friederike Böhm
- Department of Biology, Institute of Cell Biology, ETH Zurich, Zurich, Switzerland
| | | | | | | |
Collapse
|
49
|
Jahchan NS, Luo K. SnoN in mammalian development, function and diseases. Curr Opin Pharmacol 2010; 10:670-5. [PMID: 20822955 DOI: 10.1016/j.coph.2010.08.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 08/10/2010] [Accepted: 08/11/2010] [Indexed: 10/19/2022]
Abstract
SnoN (Ski-novel protein) was discovered as a nuclear proto-oncogene on the basis of its ability to induce transformation of chicken and quail embryonic fibroblasts. As a crucial negative regulator of transforming growth factor-β (TGF-β) signaling and also an activator of p53, it plays an important role in regulating cell proliferation, senescence, apoptosis, and differentiation. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed important functions of SnoN in normal epithelial development and tumorigenesis. Evidence suggests that SnoN has both pro-oncogenic and anti-oncogenic functions by modulating multiple signaling pathways. These studies suggest that SnoN may have broad functions in the development and homeostasis of embryonic and postnatal tissues.
Collapse
Affiliation(s)
- Nadine S Jahchan
- Department of Molecular and Cell Biology, University of California, Berkeley, USA
| | | |
Collapse
|
50
|
Thenappan A, Li Y, Kitisin K, Rashid A, Shetty K, Johnson L, Mishra L. Role of transforming growth factor beta signaling and expansion of progenitor cells in regenerating liver. Hepatology 2010; 51:1373-82. [PMID: 20131405 PMCID: PMC3001243 DOI: 10.1002/hep.23449] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED Adult hepatic progenitor cells are activated during regeneration when hepatocytes and bile duct epithelium are damaged or unable to proliferate. On the basis of its role as a tumor suppressor and in the potential malignant transformation of stem cells in hepatocellular carcinoma, we investigated the role of key transforming growth factor beta (TGF-beta) signaling components, including the Smad3 adaptor protein beta2-Spectrin (beta2SP), in liver regeneration. We demonstrate a streaming hepatocyte-specific dedifferentiation process in regenerating adult human liver less than 6 weeks following living donor transplantation. We then demonstrate a spatial and temporal expansion of TGF-beta signaling components, especially beta2SP, from the periportal to the pericentral zone as regeneration nears termination via immunohistochemical analysis. This expansion is associated with an expanded remaining pool of octamer 3/4 (Oct3/4)-positive progenitor cells localized to the portal tract in adult human liver from more than 6 weeks posttransplant. Furthermore, disruption of TGF-beta signaling as in the beta2SP (beta2SP+/-) knockout mouse demonstrated a striking 2 to 4-fold (P < 0.05) expanded population of Oct3/4-positive cells with activated Wnt signaling occupying an alpha-fetoprotein (AFP)+/cytokeratin-19 (CK-19)-positive progenitor cell niche following two-thirds partial hepatectomy. CONCLUSION TGF-beta signaling, particularly beta2SP, plays a critical role in hepatocyte proliferation and transitional phenotype and its loss is associated with activation of hepatic progenitor cells secondary to delayed mitogenesis and activated Wnt signaling.
Collapse
Affiliation(s)
- Arun Thenappan
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Ying Li
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Krit Kitisin
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Asif Rashid
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kirti Shetty
- Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington DC
| | - Lynt Johnson
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC,Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington DC
| | - Lopa Mishra
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC,Department of Veterans Affairs Medical Center, Washington DC
| |
Collapse
|