Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int Immunopharmacol ; 136: 112278, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38815353

ABSTRACT

Intestinal stem cells (ISCs) are pivotal for the maintenance and regeneration of the intestinal epithelium. Berberine (BBR) exhibits diverse biological activities, but it remains unclear whether BBR can modulate ISCs' function. Therefore, we investigated the effects of BBR on ISCs in healthy and radiation-injured mice and explored the potential underlying mechanisms involved. The results showed that BBR significantly increased the length of the small intestines, the height of the villi, and the depth and density of the crypts, promoted the proliferation of cryptal epithelial cells and increased the number of OLFM4+ ISCs and goblet cells. Crypts from the BBR-treated mice were more capable of growing into enteroids than those from untreated mice. BBR alleviated WAI-induced intestinal injury. BBR suppressed the apoptosis of crypt epithelial cells, increased the quantity of goblet cells, and increased the quantity of OLFM4+ ISCs and tdTomato+ progenies of ISCs after 8 Gy WAI-induced injury. Mechanistically, BBR treatment caused a significant increase in the quantity of p-S6, p-STAT3 and p-ERK1/2 positive cryptal epithelial cells under physiological conditions and after WAI-induced injury. In conclusion, BBR is capable of enhancing the function of ISCs either physiologically or after radiation-induced injury, indicating that BBR has potential value in the treatment of radiation-induced intestinal injury.


Subject(s)
Berberine , Intestinal Mucosa , Mice, Inbred C57BL , Stem Cells , Animals , Berberine/pharmacology , Berberine/therapeutic use , Stem Cells/drug effects , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Intestinal Mucosa/pathology , Male , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Goblet Cells/drug effects , Goblet Cells/radiation effects , Goblet Cells/pathology , Radiation Injuries/drug therapy , Radiation Injuries/pathology , STAT3 Transcription Factor/metabolism , Intestine, Small/drug effects , Intestine, Small/radiation effects , Intestine, Small/pathology , Intestine, Small/injuries , Intestines/drug effects , Intestines/radiation effects
2.
Int Immunopharmacol ; 131: 111908, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38518594

ABSTRACT

Radiation exposure often leads to serious health problems in humans. The intestinal epithelium is sensitive to radiation damage, and radiation causes destruction of the intestinal epithelial barrier, which leads to radiation enteritis (RE), the loss of fluids, and the translocation of intestinal bacteria and toxins; radiation can even threaten survival. In this study, we aimed to explore the influence of IVIg on the integrity of the intestinal epithelial barrier after RE. Using a RE mouse model, we investigated the protective effects of intravenous immunoglobulin (IVIg) on the epithelial junctions of RE mice and validated these findings with intestinal organoids cultured in vitro. In addition, transmission electron microscopy (TEM), western blotting (WB) and immunostaining were used to further investigate changes in intestinal epithelial ferroptosis and related signaling pathways. When RE occurs, the intestinal epithelial barrier is severely damaged. IVIg treatment significantly ameliorated this damage to epithelial tight junctions both in vivo and in vitro. Notably, IVIg alleviated RE by inhibiting intestinal epithelial ferroptosis in RE mice. Mechanistically, IVIg promoted activation of the mTOR pathway and inhibited ferroptosis in the intestinal epithelium of mice. Rapamycin, which is a potent inhibitor of the mTOR protein, significantly abolished the protective effect of IVIg against radiation-induced damage to intestinal epithelial tight junctions. Overall, IVIg can prevent RE-induced damage to the intestinal epithelial barrier and inhibit ferroptosis by activating the mTOR pathway; this study provides a new treatment strategy for patients with RE caused by radiotherapy or accidental nuclear exposure.


Subject(s)
Enteritis , Ferroptosis , Radiation Exposure , Humans , Mice , Animals , Immunoglobulins, Intravenous/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Intestines , Intestinal Mucosa , TOR Serine-Threonine Kinases/metabolism
3.
J Transl Med ; 21(1): 852, 2023 11 25.
Article in English | MEDLINE | ID: mdl-38007452

ABSTRACT

BACKGROUND: Mammalian intestinal epithelium constantly undergoes rapid self-renewal and regeneration sustained by intestinal stem cells (ISCs) within crypts. Inducible nitric oxide synthase (iNOS) is an important regulator in tissue homeostasis and inflammation. However, the functions of iNOS on ISCs have not been clarified. Here, we aimed to investigate the expression pattern of inducible nitric oxide synthase (iNOS) within crypts and explore its function in the homeostatic maintenance of the ISC niche. METHODS: Expression of iNOS was determined by tissue staining and qPCR. iNOS-/- and Lgr5 transgenic mice were used to explore the influence of iNOS ablation on ISC proliferation and differentiation. Enteroids were cultured to study the effect of iNOS on ISCs in vitro. Ileum samples from wild-type and iNOS-/- mice were collected for RNA-Seq to explore the molecular mechanisms by which iNOS regulates ISCs. RESULTS: iNOS was physiologically expressed in Paneth cells. Knockout of iNOS led to apparent morphological changes in the intestine, including a decrease in the small intestine length and in the heights of both villi and crypts. Knockout of iNOS decreased the number of Ki67+ or BrdU+ proliferative cells in crypts. Loss of iNOS increased the number of Olfm4+ ISCs but inhibited the differentiation and migration of Lgr5+ ISCs in vivo. iNOS depletion also inhibited enteroid formation and the budding efficiency of crypts in vitro. Moreover, iNOS deficiency altered gluconeogenesis and the adaptive immune response in the ileum transcriptome. CONCLUSION: Paneth cell-derived iNOS is required to maintain a healthy ISC niche, and Knockout of iNOS hinders ISC function in mice. Therefore, iNOS represents a potential target for the development of new drugs and other therapeutic interventions for intestinal disorders.


Subject(s)
Paneth Cells , Stem Cell Niche , Animals , Mice , Homeostasis , Intestinal Mucosa/metabolism , Intestines , Mammals/metabolism , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II/metabolism , Paneth Cells/metabolism , Receptors, G-Protein-Coupled/metabolism
4.
Front Pharmacol ; 14: 1332567, 2023.
Article in English | MEDLINE | ID: mdl-38273825

ABSTRACT

Mitochondria are critical for cellular energetic metabolism, intracellular signaling orchestration and programmed death regulation. Therefore, mitochondrial dysfunction is associated with various pathogeneses. The maintenance of mitochondrial homeostasis and functional recovery after injury are coordinated by mitochondrial biogenesis, dynamics and autophagy, which are collectively referred to as mitochondrial quality control. There is increasing evidence that mitochondria are important targets for melatonin to exert protective effects under pathological conditions. Melatonin, an evolutionarily conserved tryptophan metabolite, can be synthesized, transported and metabolized in mitochondria. In this review, we summarize the important role of melatonin in the damaged mitochondria elimination and mitochondrial energy supply recovery by regulating mitochondrial quality control, which may provide new strategies for clinical treatment of mitochondria-related diseases.

5.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166125, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33722746

ABSTRACT

The uncontrolled abnormal intestinal immune responses play important role in eliciting inflammatory bowel disease (IBD), yet the molecular events regulating intestinal inflammation during IBD remain poorly understood. Here, we describe an endogenous, homeostatic pattern that controls inflammatory responses in experimental murine colitis. We show that Spink7 (serine peptidase inhibitor, kazal type 7), the ortholog of human SPINK7, is significantly upregulated in dextran sodium sulfate (DSS)-induced murine colitis model. Spink7-deficient mice showed highly susceptible to experimental colitis characterized by enhanced weight loss, shorter colon length, higher disease activity index and increased colonic tissue destruction. Bone marrow reconstitution experiments demonstrated that expression of Spink7 in the immune compartment makes main contribution to its protective role in colitis. What's more, neutrophils are the primary sources of Spink7 in experimental murine colitis. Loss of Spink7 leads to augmented productions of multiple chemokines and cytokines in colitis. In summary, this study identifies neutrophils-derived endogenous Spink7-mediated control of chemokines/cytokines production as a molecular mechanism contributing to inflammation resolution during colitis.


Subject(s)
Chemokines/metabolism , Colitis/prevention & control , Cytokines/metabolism , Dextran Sulfate/toxicity , Neutrophils/metabolism , Serine Peptidase Inhibitors, Kazal Type/physiology , Serine Proteinase Inhibitors/pharmacology , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Sci Adv ; 7(1)2021 01.
Article in English | MEDLINE | ID: mdl-33523835

ABSTRACT

Acute myeloid leukemia (AML) is a deadly hematological malignancy with frequent disease relapse. The biggest challenge for AML therapy is the lack of methods to target and kill the heterogeneous leukemia cells, which lead to disease relapse. Here, we describe a near-infrared (NIR) fluorescent dye, IR-26, which preferentially accumulates in the mitochondria of AML cells, depending on the hyperactive glycolysis of malignant cell, and simultaneously impairs oxidative phosphorylation (OXPHOS) to exert targeted therapeutic effects for AML cells. In particular, IR-26 also exhibits potential for real-time monitoring of AML cells with an in vivo flow cytometry (IVFC) system. Therefore, IR-26 represents a novel all-in-one agent for the integration of AML targeting, detection, and therapy, which may help to monitor disease progression and treatment responses, prevent unnecessary delays in administering upfront therapy, and improve therapeutic efficiency to the residual AML cells, which are responsible for disease relapse.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Antineoplastic Agents/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mitochondria/metabolism , Oxidative Phosphorylation , Recurrence
7.
Br J Pharmacol ; 177(17): 3991-4006, 2020 09.
Article in English | MEDLINE | ID: mdl-32472692

ABSTRACT

BACKGROUND AND PURPOSE: There is an urgent but unmet need for mitigating radiation-induced intestinal toxicity while radio sensitising tumours for abdominal radiotherapy. We aimed to investigate the effects of metformin on radiation-induced intestinal toxicity and radiosensitivity of colorectal tumours. EXPERIMENTAL APPROACH: Acute and chronic histological injuries of the intestine from mice were used to assess radioprotection and IEC-6 cell line was used to investigate the mechanisms in vitro. The fractionated abdominal radiation model of HCT116 and HT29 tumour grafts was used to determine the effects on colorectal cancer. KEY RESULTS: Metformin alleviated radiation-induced acute and chronic intestinal toxicity by optimising mitophagy which was AMPK-dependent. In addition, our data indicated that metformin increased the radiosensitivity of colorectal tumours with P53 mutation both in vitro and in vivo. CONCLUSION AND IMPLICATIONS: Metformin may be a radiotherapy adjuvant agent for colorectal cancers especially those carrying P53 mutation. Our findings provide a new strategy for further precise clinical trials for metformin on radiotherapy.


Subject(s)
Colorectal Neoplasms , Metformin , Animals , Apoptosis , Autophagy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/radiotherapy , Metformin/pharmacology , Mice , Mutation , Tumor Suppressor Protein p53/genetics
8.
Nat Commun ; 10(1): 2538, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182708

ABSTRACT

The pathological mechanisms of radiation ulcer remain unsolved and there is currently no effective medicine. Here, we demonstrate that persistent DNA damage foci and cell senescence are involved in radiation ulcer development. Further more, we identify cordycepin, a natural nucleoside analogue, as a potent drug to block radiation ulcer (skin, intestine, tongue) in rats/mice by preventing cell senescence through the increase of NRF2 nuclear expression (the assay used is mainly on skin). Finally, cordycepin is also revealed to activate AMPK by binding with the α1 and γ1 subunit near the autoinhibitory domain of AMPK, then promotes p62-dependent autophagic degradation of Keap1, to induce NRF2 dissociate from Keap1 and translocate to the nucleus. Taken together, our findings identify cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents, and activation of AMPK or NRF2 may thus represent therapeutic targets for preventing cell senescence and radiation ulcer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cellular Senescence/drug effects , DNA Damage/drug effects , Deoxyadenosines/pharmacology , NF-E2-Related Factor 2/metabolism , Radiation Injuries, Experimental/prevention & control , Ulcer/prevention & control , Animals , Apoptosis , Cell Line , Cellular Senescence/radiation effects , DNA Damage/radiation effects , Deoxyadenosines/toxicity , Fibroblasts , Humans , Male , Mice, Inbred C57BL , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology , Rats, Sprague-Dawley , Ulcer/drug therapy , Ulcer/pathology , X-Rays/adverse effects
9.
Adv Mater ; : e1800475, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29961960

ABSTRACT

The endoplasmic reticulum (ER) stress signaling or unfolded protein response (UPR) is a common feature of many human diseases, including cancer. Excessive activation of ER stress directly induces cell death, holding a new promising strategy for the therapeutic intervention of cancer. Current ER-stress-inducing agents mainly target UPR components or proteasomes, which exert limited treatment efficacy and undesired side effects due to unselective ER stress and poor tumor-specific distribution. In this study, a unique near-infrared (NIR) fluorophore, IR-34, is synthesized and identified to selectively and efficiently trigger tumoricidal ER stress by targeting the mitochondrial protein NDUFS1. IR-34 is demonstrated to specifically accumulate in living cancer cells for tumor NIR imaging and drastically inhibit tumor growth and recurrence without causing apparent toxicity. Thus, this multifunctional NIR fluorophore may represent a novel theranostic agent for tumor imaging-guided treatment and also strengthens the idea that mitochondria could be a useful target for therapeutic ER stress in cancer cells.

10.
Adv Sci (Weinh) ; 5(3): 1700392, 2018 03.
Article in English | MEDLINE | ID: mdl-29593950

ABSTRACT

The characterization of cancer stem-like cells (CSCs) has profound implications for elucidating cancer biology and developing treatment strategies. Although surface markers are already used to identify CSCs, the expression of these markers is controversially linked to the phenotypes in different types of tumors and does not represent all functionally relevant of CSCs. Very recently, hyperactive HIF-1α/glycolysis metabolic pathway is recognized as a master regulator of CSCs. In this study, a near-infrared fluorescent small-molecule, IR-780, is identified for the exclusive characterization of human CSCs through the HIF-1α/glycolysis dependent mitochondrial transporter ABCB10's activity. The results identified for the first time that ABCB10 is involved in the preferential uptake of IR-780 in CSCs, which is regulated by HIF-1α via the direct interaction with the binding site of ABCB10 gene promoter region. In addition, IR-780 is demonstrated to conjugate with anticancer drug 5-fluorouracil to act as a potential drug delivery carrier for CSC-targeted therapy. Thus, the studies provide a new rational approach independent of surface markers to characterize CSCs via small-molecule-based imaging of HIF-1α/glycolysis hyperactive metabolic pathway dependent mitochondrial transporter's activity, which holds promise for the further development of CSCs targeted diagnostic and therapeutic strategies.

11.
EMBO J ; 37(7)2018 04 03.
Article in English | MEDLINE | ID: mdl-29467218

ABSTRACT

In 1900, Adami speculated that a sequence of context-independent energetic and structural changes governed the reversion of differentiated cells to a proliferative, regenerative state. Accordingly, we show here that differentiated cells in diverse organs become proliferative via a shared program. Metaplasia-inducing injury caused both gastric chief and pancreatic acinar cells to decrease mTORC1 activity and massively upregulate lysosomes/autophagosomes; then increase damage associated metaplastic genes such as Sox9; and finally reactivate mTORC1 and re-enter the cell cycle. Blocking mTORC1 permitted autophagy and metaplastic gene induction but blocked cell cycle re-entry at S-phase. In kidney and liver regeneration and in human gastric metaplasia, mTORC1 also correlated with proliferation. In lysosome-defective Gnptab-/- mice, both metaplasia-associated gene expression changes and mTORC1-mediated proliferation were deficient in pancreas and stomach. Our findings indicate differentiated cells become proliferative using a sequential program with intervening checkpoints: (i) differentiated cell structure degradation; (ii) metaplasia- or progenitor-associated gene induction; (iii) cell cycle re-entry. We propose this program, which we term "paligenosis", is a fundamental process, like apoptosis, available to differentiated cells to fuel regeneration following injury.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Regeneration/physiology , Acinar Cells , Animals , Autophagosomes/physiology , Cell Cycle/physiology , Cell Transdifferentiation/physiology , Cellular Reprogramming/physiology , Chief Cells, Gastric/pathology , Gastrointestinal Tract/pathology , Gene Expression , Humans , Lysosomes , Metaplasia/genetics , Mice , Mice, Inbred C57BL , S Phase/physiology , SOX9 Transcription Factor/metabolism , Stomach/injuries , Stomach/pathology , Transferases (Other Substituted Phosphate Groups)/genetics
12.
Article in English | MEDLINE | ID: mdl-29158275

ABSTRACT

The increasing incidence of multidrug-resistant Acinetobacter baumannii (MDRAb) infections worldwide has necessitated the development of novel antibiotics. Human defensin 5 (HD5) is an endogenous peptide with a complex architecture and antibacterial activity against MDRAb In the present study, we attempted to simplify the structure of HD5 by removing disulfide bonds. We found that the Cys2-4 bond was most indispensable for HD5 to inactivate MDRAb, although the antibacterial activity of the derivative was significantly attenuated. We then replaced the noncationic and nonhydrophobic residues with electropositive Arg to increase the antibacterial activity of HD5 derivative that contains a Cys2-4 bond, obtaining another derivative termed HD5d5. The in vitro antibacterial assay and irradiation-wound-infection animal experiment both showed that HD5d5 was much more effective than HD5 at eliminating MDRAb Further investigations revealed that HD5d5 efficiently bound to outer membrane lipid A and penetrated membranes, leading to bacterial collapse and peptide translocation. Compared to HD5, more HD5d5 molecules were located in the cytoplasm of MDRAb, and HD5d5 was more efficient at reducing the activities of superoxide dismutase and catalase, causing the accumulation of reactive oxygen species that are detrimental to microbes. In addition, HD5 failed to suppress the pathogenic outer membrane protein A of Acinetobacter baumannii (AbOmpA) at concentrations up to 50 µg/ml, whereas HD5d5 strongly bound to AbOmpA and exhibited a dramatic toxin-neutralizing ability, thus expanding the repertoire of drugs that is available to treat MDRAb infections.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial , Wound Infection/drug therapy , alpha-Defensins/pharmacology , Acinetobacter Infections/microbiology , Acinetobacter Infections/mortality , Acinetobacter Infections/pathology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/metabolism , Animals , Anti-Bacterial Agents/chemical synthesis , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalase/antagonists & inhibitors , Catalase/genetics , Catalase/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Lipid A/metabolism , Mice , Mice, Inbred BALB C , Protein Binding , Protein Engineering/methods , Protein Isoforms/chemical synthesis , Protein Isoforms/pharmacology , Protein Transport , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Survival Analysis , Whole-Body Irradiation , Wound Infection/microbiology , Wound Infection/mortality , Wound Infection/pathology , alpha-Defensins/chemical synthesis
13.
Genes Dev ; 31(2): 154-171, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28174210

ABSTRACT

We hypothesized that basic helix-loop-helix (bHLH) MIST1 (BHLHA15) is a "scaling factor" that universally establishes secretory morphology in cells that perform regulated secretion. Here, we show that targeted deletion of MIST1 caused dismantling of the secretory apparatus of diverse exocrine cells. Parietal cells (PCs), whose function is to pump acid into the stomach, normally lack MIST1 and do not perform regulated secretion. Forced expression of MIST1 in PCs caused them to expand their apical cytoplasm, rearrange mitochondrial/lysosome trafficking, and generate large secretory granules. Mist1 induced a cohort of genes regulated by MIST1 in multiple organs but did not affect PC function. MIST1 bound CATATG/CAGCTG E boxes in the first intron of genes that regulate autophagosome/lysosomal degradation, mitochondrial trafficking, and amino acid metabolism. Similar alterations in cell architecture and gene expression were also caused by ectopically inducing MIST1 in vivo in hepatocytes. Thus, MIST1 is a scaling factor necessary and sufficient by itself to induce and maintain secretory cell architecture. Our results indicate that, whereas mature cell types in each organ may have unique developmental origins, cells performing similar physiological functions throughout the body share similar transcription factor-mediated architectural "blueprints."


Subject(s)
Gene Expression Regulation/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Parietal Cells, Gastric/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Secretory Pathway/genetics , Acinar Cells/cytology , Acinar Cells/drug effects , Acinar Cells/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cell Line , Ectopic Gene Expression/drug effects , Gene Deletion , Gene Expression Regulation/drug effects , Mice , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/ultrastructure , Tamoxifen/pharmacology
14.
Gastroenterology ; 152(4): 762-766.e7, 2017 03.
Article in English | MEDLINE | ID: mdl-27932312

ABSTRACT

Parietal cell atrophy is considered to cause metaplasia in the stomach. We developed mice that express the diphtheria toxin receptor specifically in parietal cells to induce their death, and found this to increase proliferation in the normal stem cell zone and neck but not to cause metaplastic reprogramming of chief cells. Furthermore, the metaplasia-inducing agents tamoxifen or DMP-777 still induced metaplasia even after previous destruction of parietal cells by diphtheria toxin. Atrophy of parietal cells alone therefore is not sufficient to induce metaplasia: completion of metaplastic reprogramming of chief cells requires mechanisms beyond parietal cell injury or death.


Subject(s)
Apoptosis , Chief Cells, Gastric/pathology , Parietal Cells, Gastric/pathology , Parietal Cells, Gastric/physiology , Stomach/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Atrophy/chemically induced , Azetidines , Cell Proliferation , Cellular Reprogramming , Chief Cells, Gastric/metabolism , Diphtheria Toxin/pharmacology , Heparin-binding EGF-like Growth Factor/genetics , Intercellular Signaling Peptides and Proteins , Intrinsic Factor/metabolism , Metaplasia/chemically induced , Metaplasia/genetics , Metaplasia/metabolism , Mice , Parietal Cells, Gastric/drug effects , Peptides/metabolism , Piperazines , Plant Lectins/metabolism , Tamoxifen
15.
Sci Rep ; 6: 25314, 2016 04 29.
Article in English | MEDLINE | ID: mdl-27126764

ABSTRACT

Progressive liver disease is a major health issue for which no effective treatment is available, leading to cirrhosis and orthotopic liver transplantation. However, the lack of availability of donor organs and other adverse factors including rejection limit its extensive clinical application. Cell-based therapy using mesenchymal stem/stromal cells (MSCs) may represent an attractive therapeutic option. Dermal-derived mesenchymal cells (DMCs) are attractive as one of the abundant sources from which to isolate mesenchymal cells for therapeutic applications and can be easily accessed with minimal harm to the donor. In this study, we used two different animal models to investigate potential therapeutic effect of DMCs transplantation in liver injury. We found that DMCs administration alleviated liver fibrosis and restored the liver function in fibrotic mice induced by CCl4. Furthermore, in an acute irradiation induced damage model, a unique population of DMCs could engraft into the liver tissue for a long period, exhibiting the phenotype of both mesenchymal cells and macrophage cells, and improve the survival of mice exposed to 8 Gy lethally total-body irradiation. These discoveries provide important evidence that DMCs therapy has a beneficial effect on liver injury, and provide new insight into liver injury therapy depending on the alternative cells.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Liver Regeneration , Mesenchymal Stem Cell Transplantation , Radiation Injuries, Experimental/therapy , Animals , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Liver Function Tests , Mice , Radiation Injuries, Experimental/pathology , Skin/cytology , Treatment Outcome
16.
Mol Cell Biochem ; 378(1-2): 107-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23456479

ABSTRACT

Although mesenchymal stem cells (MSCs) promote lung cancer growth in vivo, in vitro studies indicate that they inhibit the proliferation of lung cancer cells. Because malignant tumors contain a heterogeneous cell population with variable capacity for self-renewal, the aim of this study was to determine whether the inconsistencies between in vitro and in vivo studies are a result of differential effects of MSCs on the heterogeneous cell population within lung cancer cell lines. Human MSCs were isolated from the bone marrow, and their cell surface antigen expression and multi-lineage differentiation capacity was examined at passage 10. CD133+ cells were isolated from A549 and H446 cell lines using immunomagnetic separation. The effects of MSCs on the growth and microsphere formation of heterogeneous cell populations within two lung cancer cell lines (A549 and H446) were compared. MSCs inhibited the in vitro proliferation of both cell lines, but significantly accelerated tumor formation and stimulated tumor growth in vivo (P < 0.05). In CD133+ cells isolated from both A549 and H446 cells, co-culture with MSCs for 1-3 days significantly increased their proliferation (P < 0.05). MSCs also significantly increased microsphere formation in both cell lines (P < 0.05). Selective stimulation of CD133+ cell growth may account for the discrepant effects of MSCs on lung cancer progression.


Subject(s)
Lung Neoplasms/pathology , Mesenchymal Stem Cells/physiology , Neoplastic Stem Cells/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Female , Glycoproteins/metabolism , Humans , Immunomagnetic Separation , Mice , Mice, Nude , Neoplasm Transplantation , Peptides/metabolism
17.
J Surg Res ; 183(1): 427-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23462453

ABSTRACT

BACKGROUND: Bone marrow-derived mesenchymal stem cells (BM-MSCs) play a crucial role in tissue repair. Their role in thermal burn wound regeneration and the relevant mechanism, however, is rarely studied. METHODS: BM-MSCs from green fluorescent protein transgenic male mice were transfused to irradiated recipient female C57BL/6 mice. Twenty-one days later, the female mice were inflicted with burn wounds. The size of the burned area was measured by an in vivo fluorescence imaging system, and BM-MSC chemotaxis and epithelialization were estimated by fluorescence in situ hybridization and immunofluorescence technology. The expression of CXCL12 and CXCR4 in the wound margin was detected by enzyme-linked immunosorbent assay and immunohistochemistry. The importance of CXCL12/CXCR4 signaling in BM-MSC chemotaxis was further estimated by blocking CXCR4 in vivo and in vitro. RESULTS: In vivo imaging results showed that BM-MSCs migrated to the injured margins. Fluorescence in situ hybridization and immunofluorescence technology revealed that Y chromosome-positive cells derived from green fluorescent protein transgenic mice were detected to be colocalized with keratin protein. Enzyme-linked immunosorbent assay revealed increased levels of CXCL12 and CXCR4 protein in the wound sites of BM-MSC-treated chimeric mice after burn. Immunohistochemistry also disclosed that CXCL12 levels were elevated at postburn day 7 compared with day 0. Furthermore, pretreatment of the BM-MSCs with the CXCR4 antagonist AMD3100 significantly inhibited the mobilization of BM-MSCs in vitro and in vivo, which attenuated wound closure. CONCLUSION: BM-MSC migration to the burned margins promotes the epithelialization of the wound, and mobilization of BM-MSCs is mediated by CXCL12/CXCR4 signaling.


Subject(s)
Burns/metabolism , Chemokine CXCL12/metabolism , Mesenchymal Stem Cells/physiology , Re-Epithelialization , Receptors, CXCR4/metabolism , Animals , Chemotaxis , Chimera , Epidermal Cells , Female , Green Fluorescent Proteins , Hair Follicle/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Tumour Biol ; 34(1): 145-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23055190

ABSTRACT

Bone marrow-derived stem cells (BMDCs) have the ability to differentiate into lung epithelial cells in response to damage; however, their role in squamous cell carcinoma (SCC) formation is unknown. This study aimed to determine whether BMDC-derived lung epithelial cells could contribute to SCC formation. A model of lung SCC induced with N-nitrosodiethylamine (NDEA) in recipient female mice transplanted with green fluorescent protein (GFP)-positive BMDCs from male donors was established. Incorporation of BMDCs in lung tissue was determined using immunohistochemistry and immunofluorescence to detect GFP expression and fluorescence in situ hybridization to Y chromosomes. BMDC appeared at three stages of lung SCC progression: metaplasia, dysplasia, and carcinoma. There was a significantly higher proportion of GFP-positive (GFP(+)) cells within SCC than was found in metaplasia and dysplasia 16 weeks post-transplantation (both P < 0.017); GFP(+) BMDCs were also observed in clusters within several SCC nests. Furthermore, most GFP(+) cells in SCC were pancytokeratin-positive (PCK(+)) epithelial cells, and some exhibited proliferative activity as determined by Ki67 staining (9.7 ± 3.92 %). The presence of GFP(+)Ki67(+)PCK(+) cells within SCC nests suggested that some donor BMDCs differentiated into proliferating epithelial cells. Finally, analysis of p63 expression, a marker of SCC cells, indicated that the presence of GFP(+)p63(+) cells (green) in inner parts of the SCC. These findings strongly suggest that BMDC-derived lung epithelial cells could participate in lung SCC formation and partially contribute to tumor growth, which might have significant potential implications for both clinical cancer therapy using BMDCs.


Subject(s)
Bone Marrow Cells/physiology , Carcinoma, Squamous Cell/pathology , Epithelial Cells/physiology , Lung Neoplasms/pathology , Stem Cells/physiology , Animals , Bone Marrow Transplantation , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Diethylnitrosamine , Female , Green Fluorescent Proteins , Keratinocytes/pathology , Keratins/biosynthesis , Ki-67 Antigen/analysis , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphoproteins/analysis , Trans-Activators/analysis
19.
Mol Cell Biochem ; 364(1-2): 29-37, 2012 May.
Article in English | MEDLINE | ID: mdl-22198336

ABSTRACT

Steroid receptor coactivator-3 (SRC-3) is a multifunctional protein that plays an important role in mammary gland growth, development, and tumorigenesis. In this study, SCR-3 gene knockout mice were used to study the effects of SCR-3 on the immunosuppression accompanied with systemic inflammatory response syndrome (SIRS). Bacterial clearance assay was performed by blood culture and frozen sections, and the results showed that the absence of SCR-3 protein serious damaged the innate immune system and the body's ability to inactivate or phagocytosis of bacteria was significantly decreased, and the absence of SCR-3 protein also weakened phagocytes' ability to degrade bacteria and their metabolites. Furthermore, animal model of inflammatory reaction was established and the immune function was determined, and the results revealed that SRC-3 protein may play an important role in maintenance of T-cells' immune function, and severe T-cell immune function disorder would be resulted once SRC-3 protein is missing. In addition, the results of our study showed the steady-state of lymphocyte subsets was destroyed after SIRS, leading the suppression of cellular immune function, and the absence of SCR-3 protein may aggravate the suppression of T-lymphocyte function. Therefore, the present study demonstrated that the absence of SCR-3 protein would aggravate immunosuppression. In addition, SRC-3 protein is a significant regulator of infection and inflammation, and SRC-3 protein play an essential role in the development of immunosuppression accompanied with SIRS.


Subject(s)
Immunity, Innate , Nuclear Receptor Coactivator 3/immunology , Systemic Inflammatory Response Syndrome/immunology , T-Lymphocytes/immunology , Animals , Bacteria/pathogenicity , Female , Immunity, Innate/genetics , Interleukin-2/blood , Lipopolysaccharides/administration & dosage , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Nuclear Receptor Coactivator 3/deficiency , Nuclear Receptor Coactivator 3/genetics , Phagocytosis/genetics , Systemic Inflammatory Response Syndrome/blood , Systemic Inflammatory Response Syndrome/genetics , T-Lymphocytes/pathology
20.
Cell Transplant ; 20(5): 741-51, 2011.
Article in English | MEDLINE | ID: mdl-21054944

ABSTRACT

Near-infrared (NIR) fluorescent agents hold great promise for noninvasive in vivo imaging. We have recently reported that a NIR fluorescent heptamethine dye, IR-780 iodide, exhibits unique optical properties for biomedical imaging. On the basis of this foregoing work, we further describe here the potential application of IR-780 iodide as a novel NIR agent for stem cell labeling and tracking. The labeling efficiency, subcellular localization, and the effects on cell viability and differentiation of IR-780 iodide were investigated. The in vivo distribution of stem cells after intravenous transplantation was traced by whole-body animal NIR imaging. Our results showed that IR-780 iodide exhibited superior labeling efficiency and biocompatibility with unique optical properties. Following whole-body NIR imaging, the pulmonary passage of stem cells was noninvasively visualized in rats after systemic transplantation of IR-780 iodide-labeled stem cells through intravenous delivery. With this NIR imaging method, we further confirmed that pretreatment with sodium nitroprusside (SNP), a vasodilator agent, significantly reduced the cell trapping in the lung and increased the cell passage through the lung capillaries. Our study suggests that IR-780 iodide may represent an effective NIR fluorophore for stem cell labeling and tracking.


Subject(s)
Fluorescent Dyes/chemistry , Indoles/chemistry , Spectroscopy, Near-Infrared , Stem Cells/cytology , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Nitroprusside/chemistry , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation , Stem Cells/chemistry , Whole Body Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...