Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 525, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702433

ABSTRACT

Disabled 2 (Dab2), an adaptor protein, is up regulated in the hair follicle stem cells (HFSCs); however, its role in any tissue stem cells has not been studied. In the present study, we have reported that Dab2 conditional knockout (Dab2-cKO) mice exhibited a delay in the HF cycle due to perturbed activation of HFSCs. Further, Dab2-cKO mice showed a reduction in the number of HFSCs and reduced colony forming ability of HFSCs. Dab2-cKO mice showed extended quiescence of HFSCs concomitant with an increased expression of Nfatc1. Dab2-cKO mice showed a decreased expression of anti-aging genes such as Col17a1, decorin, Sirt2 and Sirt7. Dab2-cKO mice did not show full hair coat recovery in aged mice thereby suggesting an accelerated aging process. Overall, we unveil for the first time, the role of Dab2 that regulate activation and self-renewal of HFSCs.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Hair Follicle , Mice, Knockout , Stem Cells , Animals , Hair Follicle/metabolism , Hair Follicle/cytology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Stem Cells/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Cell Self Renewal/genetics , Mice, Inbred C57BL , Cell Proliferation
2.
Biochem Biophys Res Commun ; 677: 98-104, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37566923

ABSTRACT

Breast cancer is the second most cancer worldwide in females. The primary factor responsible for tumor recurrence is the presence of breast cancer stem cells (BCSCs), which escape the chemo-radiotherapy. In this study, we have investigated the role of Secretory phospholipase-A2 Group 2A (sPLA2-IIA) that is overexpressed in BCSCs of MCF7 and MDA-MB-231 breast cancer cell lines. Further, overexpression of sPLA2-IIA revealed an increased EGFR/JNK/c-JUN/c-FOS signaling in BCSCs, while sPLA2-IIA knockdown significantly reduced the percentage of BCSCs and decreased signaling in both the cell lines. Importantly, sPLA2-IIA knockdown showed differentiation of BCSCs. Strikingly, PET imaging showed a decreased metastatic potential of BCSCs. Our study revealed a novel role of sPLA2-IIA in regulating BCSCs, which play a crucial role in regulating the differentiation and metastatic potential of BCSCs.


Subject(s)
Breast Neoplasms , Phospholipases A2, Secretory , Female , Humans , Phospholipases A2, Secretory/genetics , Phospholipases , Neoplasm Recurrence, Local , Cell Differentiation , Neoplastic Stem Cells , Group II Phospholipases A2/genetics
3.
Mol Biol Rep ; 50(10): 8469-8481, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37639153

ABSTRACT

BACKGROUND: Oral Squamous Cell Carcinoma (OSCC) is a highly prevalent cancer in the Indian subcontinent. The major cause of mortality in OSCC patients is metastasis. Epithelial-to-mesenchymal transition (EMT) marks an important step in the metastatic process. Additionally, TP53, an important tumor suppressor gene, is also a significant determinant of the treatment outcome, and also plays a role in EMT. Therefore, understanding the interconnections between ultrastructural features, EMT status and TP53 mutational status is of vital importance. METHODS AND RESULTS: The ultrastructure of five OSCC cell lines was visualized by transmission electron microscopy. Trans-well invasion and migration assays as well as scratch-wound assay, and the expression of various EMT-related genes were utilized to assess the EMT status of the cell lines. The TP53 exons were amplified for the ACOSC3, ACOSC4 and ACOSC16 cell lines and sequenced and the mutations in the gene were identified by sequence alignment. The TP53 mutation in the UPCI:SCC029B cell line has been previously reported, while UPCI:SCC040 has been reported to harbor a wild type TP53. The ACOSC4 cell line which showed the shortest intercellular gaps, also had the least invasive and migratory potential. Interestingly, ACOSC4 showed the highest expression of E-cadherin and the lowest expression of Vimentin, TWIST1, ZEB1, and MMPs. Additionally, TP53 gene of ACOSC4 was unmutated, whereas the ACOSC3 and ACOSC16 harbored TP53 mutations. The mutation in ACOSC3 (R196*) was also found in 7 TCGA samples. Similarly, the UPCI:SCC040 cell line that harbors a wild type TP53 showed shorter intracellular gaps. CONCLUSIONS: Cellular migratory properties are associated with cellular ultrastructure, epithelial-to-mesenchymal transition status and the status of TP53 mutation in the genome.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Mouth Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Epithelial-Mesenchymal Transition/genetics , Squamous Cell Carcinoma of Head and Neck , Cell Line , Tumor Suppressor Protein p53/genetics
4.
World J Stem Cells ; 15(5): 438-452, 2023 May 26.
Article in English | MEDLINE | ID: mdl-37342225

ABSTRACT

Head and neck squamous cell carcinoma is the seventh most common cancer worldwide with high mortality rates. Amongst oral cavity cancers, tongue carcinoma is a very common and aggressive oral cavity carcinoma. Despite the implementation of a multimodality treatment regime including surgical intervention, chemo-radiation as well as targeted therapy, tongue carcinoma shows a poor overall 5-year survival pattern, which is attributed to therapy resistance and recurrence of the disease. The presence of a rare population, i.e., cancer stem cells (CSCs) within the tumor, are involved in therapy resistance, recurrence, and distant metastasis that results in poor survival patterns. Therapeutic agents targeting CSCs have been in clinical trials, although they are unable to reach into therapy stage which is due to their failure in trials. A more detailed understanding of the CSCs is essential for identifying efficient targets. Molecular signaling pathways, which are differentially regulated in the CSCs, are one of the promising targets to manipulate the CSCs that would provide an improved outcome. In this review, we summarize the current understanding of molecular signaling associated with the maintenance and regulation of CSCs in tongue squamous cell carcinoma in order to emphasize the need of the hour to get a deeper understanding to unravel novel targets.

5.
Methods Cell Biol ; 171: 63-80, 2022.
Article in English | MEDLINE | ID: mdl-35953206

ABSTRACT

Skin squamous cell carcinoma (skin SCC) is the most frequently occurring cancer. Skin is the first line of defense that provides protection from the external environment. Skin consists of epidermis, dermis, and hypodermis. The epidermis comprises of inter-follicular epidermis, hair follicles, sebaceous glands, and sweat glands. Stem cells within these epidermal compartments play crucial role in epidermal regeneration and repair. Various factors such as higher exposure to ultraviolet light (UV) of sun, genetic predisposition, exposure to carcinogens, etc. that give rise to skin cancer. Within the skin SCC, there exists a pool of cancer stem cells (CSCs) that are highly quiescent with self-renewal capacity. Further, isolation and molecular characterization of CSCs would enable to unravel mechanism involved in tumor progression, metastasis, relapse, and resistance to chemotherapeutic agents. To understand the sequential events of carcinogenesis, the two-stage skin carcinogenesis murine model is proposed, which employs the topical application of a chemical carcinogen, DMBA that causes several activating mutations occurring in the genes responsible for cell proliferation and growth. Further, initiation is followed by tumor promotion, which is induced by repeated application of tumor-promoting agent, TPA, which fixes the activating mutations resulting in the formation of a benign papilloma. Subsequently, papilloma further progresses to highly malignant SCC. Here, using the two-stage skin carcinogenesis murine model, we provide a detailed protocol for the isolation of CSCs from murine skin SCC. FACS sorting of CSCs is followed by assays such as invitro-spheroid assay, in vivo-tumorigenesis-limiting dilution and in vivo-tumorigenesis-serial transplantation assay and expression profiling.


Subject(s)
Carcinoma, Squamous Cell , Papilloma , Skin Neoplasms , 9,10-Dimethyl-1,2-benzanthracene/adverse effects , Animals , Carcinogenesis , Carcinogens , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/genetics , Disease Models, Animal , Humans , Mice , Neoplastic Stem Cells/pathology , Papilloma/chemically induced , Papilloma/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Tetradecanoylphorbol Acetate/adverse effects
6.
Stem Cells ; 40(9): 802-817, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35689817

ABSTRACT

Wnt signaling plays a pivotal role in regulating activation, proliferation, stem cell renewal, and differentiation of hair follicle stem cells (HFSCs). Secreted frizzled-related protein 1 (Sfrp1), a Wnt antagonist is upregulated in the HFSCs; however, its role in the HFSCs regulation is still obscure. Here, we show that Sfrp1 loss showed a depletion of HFSCs, enhanced HFSC proliferation, and faster hair follicle cycle at PD21-PD28; HFSC markers, such as Lgr5 and Axin2, were decreased in both the Sfrp1+/- and Sfrp1-/- HFSCs. In addition, the second hair follicle cycle was also faster compared with WT. Importantly, Sfrp1-/- showed a restoration of HFSC by second telogen (PD49), whereas Sfrp1+/- did not show restoration with still having a decreased HFSC. In fact, restoration of HFSCs was due to a pronounced downregulation of ß-catenin activity mediated through a cross-talk of BMP-AKT-GSK3ß signaling in Sfrp1-/- compared with Sfrp1+/-, where downregulation was less pronounced. In cultured keratinocytes, Sfrp1 loss resulted in enhanced proliferation and clonogenicity, which were reversed by treating with either BMPR1A or GSK3ß inhibitor thereby confirming BMP-AKT-GSK3ß signaling involved in ß-catenin regulation in both the Sfrp1+/- and Sfrp1-/- mice. Our study reveals a novel function of Sfrp1 by unraveling an in vivo molecular mechanism that regulates the HFSCs pool mediated through a hitherto unknown cross-talk of BMP-AKT-GSK3ß signaling that maintains stem cell pool balance, which in turn maintains skin tissue homeostasis.


Subject(s)
Hair Follicle , Membrane Proteins/metabolism , beta Catenin , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
7.
Biochem Biophys Res Commun ; 597: 102-108, 2022 Mar 15.
Article in English | MEDLINE | ID: mdl-35134607

ABSTRACT

Breast cancer stem cells (BCSCs) are slow cycling cells that escape the traditional chemo-radio-therapy, thereby contributing in resistance and recurrence. Although several markers have been identified, it is still challenging to develop strategies targeting them. In this study, we have isolated BCSCs from MCF-7 cell line using markers CD44+/CD24-/low, which showed higher percentage of mammospheres in CSC population. Moreover, in vivo tumorigenic potential of BCSCs showed as low as 10,000 cells had the ability to develop tumors when transplanted into NOD-SCID mice. We observed an increased level of EMT markers in CSC population. Overexpression of secretory phospholipase sPLA2-IIA was found in CSCs. Further, we have uncovered the upregulation of sPLA2-IIA mediated through JNK signaling in breast cancer cells whereas knockdown of sPLA2-IIA reduces JNK signaling, cell proliferation, EMT and in vivo tumorigenic potential in breast cancer cells. Our study reveals overexpression of sPLA2-IIA in two different breast cancer cells such as MCF7 (ER+,PR+) and a triple negative, MDA-MB-231 (ER-PR-HER2-). Further, the novel role of sPLA2-IIA was discerned by unraveling the molecular mechanism, which regulates the cell proliferation and metastasis in breast cancer cells.

8.
J Biomol Struct Dyn ; 40(15): 6831-6844, 2022 09.
Article in English | MEDLINE | ID: mdl-33666148

ABSTRACT

cWnt-signalling plays a crucial role in stem cell maintenance and tissue homeostasis. Secreted frizzled-related proteins(SFRP), Wnt inhibitors consist of the N-terminal cysteine rich domain(CRD) and the C-terminal netrin(NTR) domain. SFRP1 binds to the Wnt ligands and frizzled receptors(FZ) either through its SFRP1CRD or through its SFRP1Netrin domains; however, very little is known on these binding affinities. Here, we attempted to understand the interactions and binding affinities of SFRP1-Wnt5B, SFRP1-FZ(2, 3 & 7) and Wnt5B-FZ(2, 3 & 7) that are mainly expressed in murine hair follicle stem cells. SFRP1CRD, SFRP1Netrin, Wnt5B and FZ(2, 3 & 7) structures were built using homology modelling, followed by their molecular dynamics simulations. SFRP1CRD showed lower fluctuation when in complex with FZ2, FZ3 and FZ7 and Wnt5B as compared to SFRP1Netrin using RMSF and RMSD. However, free energy showed SFRP1Netrin was energetically more stable than SFRP1CRD. SFRP1Netrin formed more number of interactions with FZ as compared to SFRP1CRD. Importantly, SFRP1Netrin favoured binding to the FZ receptors(FZ3 > FZ7 > FZ2) as compared to Wnt5B ligand. Conversely, the SFRP1CRD showed more affinity towards the Wnt5B ligand as compared to FZ receptors. Wnt5B showed the best binding affinity with FZ3 followed by SFRP1CRD and SFRP1Netrin. Therefore, SFRP1Netrin can bind to the FZ3 with higher binding affinity and may inhibit non-canonical Wnt-signalling pathway. Our study provides the comprehensive information on the binding affinities among the Wnt5B, SFRP1CRD/Netrin and FZ(2, 3 & 7). Thus, this information might also help in designing novel strategies to inhibit aberrant Wnt-signalling.Communicated by Ramaswamy H. Sarma.


Subject(s)
Frizzled Receptors , Wnt Proteins , Animals , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism , Ligands , Membrane Proteins , Mice , Netrins , Signal Transduction , Wnt Proteins/chemistry , Wnt Proteins/metabolism
9.
Front Biosci (Landmark Ed) ; 26(2): 238-265, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33049669

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a fundamental cellular phenomenon that plays an intrinsic role in development, tissue repair, and cancer progression. EMT is tightly regulated by transcription factors that alter gene expression to promote epithelial to mesenchymal phenotype. EMT is also regulated by a diverse array of cytokines and growth factors whose activities are deregulated during malignancy. EMT enables tumor cells to exist in various intermediate states along the epithelial-mesenchymal phenotypic axis that transit from cancer stem cells (CSCs) to circulating tumor cells (CTCs). Recent studies have revealed the importance of CSCs in tumor promotion, invasion and metastasis. The relapsed tumors encompass CSCs which are resistant to radiotherapy and chemotherapy. In this review, we have summarized our current understanding of the molecular mechanisms that regulate EMT induced CSC phenotype. We have highlighted studies implicating the function of TGF-�, Wnt, and Notch regulated non-coding RNAs in driving EMT promoting CSC self-renewal. Finally, we discuss how the EMT and CSCs cause drug resistance with the hope to overcome such resistance as a possible approach for cancer treatment.


Subject(s)
Epithelial-Mesenchymal Transition , Neoplasms/therapy , Neoplastic Stem Cells/cytology , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology
10.
Sci Rep ; 10(1): 4951, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-32170155

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Stem Cell Reports ; 14(2): 271-284, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31928951

ABSTRACT

Wnt signaling is involved in the regulation of cancer stem cells (CSCs); however, the molecular mechanism involved is still obscure. SFRP1, a Wnt inhibitor, is downregulated in various human cancers; however, its role in tumor initiation and CSC regulation remains unexplored. Here, we used a skin carcinogenesis model, which showed early tumor initiation in Sfrp1-/- (Sfrp1 knockout) mice and increased tumorigenic potential of Sfrp1-/- CSCs. Expression profiling on Sfrp1-/- CSCs showed upregulation of genes involved in epithelial to mesenchymal transition, stemness, proliferation, and metastasis. Further, SOX-2 and SFRP1 expression was validated in human skin cutaneous squamous cell carcinoma, head and neck squamous cell carcinoma, and breast cancer. The data showed downregulation of SFRP1 and upregulation of SOX-2, establishing their inverse correlation. Importantly, we broadly uncover an inverse correlation of SFRP1 and SOX-2 in epithelial cancers that may be used as a potential prognostic marker in the management of cancer.


Subject(s)
Carcinogenesis/metabolism , Epithelium/pathology , Membrane Proteins/metabolism , Neoplastic Stem Cells/pathology , Skin Neoplasms/pathology , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/deficiency , Mice , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Up-Regulation/genetics
12.
EBioMedicine ; 48: 364-376, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31521610

ABSTRACT

BACKGROUND: Tissue stem cells (SCs) and cancer cells proliferation is regulated by many common signalling mechanisms. These mechanisms temporally balance proliferation and differentiation events during normal tissue homeostasis and repair. However, the effect of these aberrant signalling mechanisms on the ultimate fate of SCs and cancer cells remains obscure. METHODS: To evaluate the functional effects of Secretory Phospholipase A2-IIA (sPLA2-IIA) induced abnormal signalling on normal SCs and cancer cells, we have used K14-sPLA2-IIA transgenic mice hair follicle stem cells (HFSCs), DMBA/TPA induced mouse skin tumour tissues, human oral squamous cell carcinoma (OSCC) and skin squamous cell carcinoma (SCC) derived cell lines. FINDINGS: Our study demonstrates that sPLA2-IIA induces rapid proliferation of HFSCs, thereby altering the proliferation dynamics leading to a complete loss of the slow cycling H2BGFP positive HFSCs. Interestingly, in vivo reversion study by JNK inhibition exhibited a significant delay in post depilation hair growth, confirming that sPLA2-IIA promotes HFSCs proliferation through JNK/c-Jun signalling. In a different cellular context, we showed increased expression of sPLA2-IIA in human OSCC and mouse skin cancer tissues. Importantly, a xenograft of sPLA2-IIA knockdown cells of OSCC and SCC cell lines showed a concomitant reduction of tumour volume in NOD-SCID mice and decreased JNK/c-Jun signalling. INTERPRETATION: This study unravels how an increased proliferation induced by a common proliferation inducer (sPLA2-IIA) alters the fate of normal SCs and cancer cells distinctively through common JNK/c-Jun signalling. Thus, sPLA2-IIA can be a potential target for various diseases including cancer. FUND: This work was partly supported by the Indian Council of Medical Research (ICMR-3097) and ACTREC (42) grants.


Subject(s)
Carcinoma/genetics , Carcinoma/metabolism , Group II Phospholipases A2/genetics , Hair Follicle/cytology , Stem Cells/metabolism , Animals , Carcinoma/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Group II Phospholipases A2/metabolism , Humans , Keratinocytes/metabolism , MAP Kinase Signaling System , Mice , Mice, Transgenic
13.
Oncol Rep ; 41(4): 2289-2298, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816493

ABSTRACT

Oral squamous cell carcinoma (OSCC) is a leading cause of mortality in India owing to the high percentage of tobacco chewers, smokers and alcohol consumption. OSCC is highly heterogeneous in nature; therefore poses a challenge in the treatment of the patient. To better understand the heterogeneity of the tumors, an in vitro cell line model is required. However, the efficiency of establishing cell lines from the oral tumors is low. In the present study, three novel cell lines, namely ACOSC3, ACOSC4, and ACOSC16, were isolated and characterized from advanced­stage treatment­naive OSCCs originating from the buccal mucosa. The three cell lines exhibited polygonal morphology, which is typical of epithelial cells. Furthermore, immunofluorescence revealed the expression of keratins 8 and 14, thereby confirming the epithelial origin of the cells. DNA content analysis of the three OSCC cell lines revealed aneuploidy. Furthermore, an in vitro orosphere assay revealed the formation of primary orospheres. Notably, the OSCC cell lines were able to give rise to tumors when administered subcutaneously into non­obese diabetic/severe combined immune deficiency mice. The novelty of the cell lines was also validated by performing short tandem repeat profiling; the STR profiles of the present cell lines did not significantly match with any known established OSCC cell lines present in the DSMZ database, thereby confirming the unique identity of these lines. These cell lines established from tumor samples derived from Indian OSCC patients provide a valuable resource to understand the molecular mechanism involved in tumor resistance and recurrence.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Mouth Neoplasms/pathology , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Mouth Mucosa/cytology , Mouth Mucosa/pathology , Primary Cell Culture , Spheroids, Cellular , Xenograft Model Antitumor Assays
14.
Int J Radiat Oncol Biol Phys ; 103(1): 161-168, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30213750

ABSTRACT

PURPOSE: Although cancer stem cells (CSCs) have been reported across solid tumors, there is a dearth of data regarding CSC and its impact on outcomes of cervical cancer. METHODS AND MATERIALS: From October 2013 to December 2015, patients with squamous cancer of the cervix (stage IB2-IVA) were included. Pretreatment and posttreatment biopsy was obtained and immunohistochemistry was performed for SOX-2, OCT-4, Nanog, CD44, and Podoplanin. All patients received concurrent radiation and brachytherapy to an equivalent dose of 80 to 84 Gy to point A with concurrent weekly cisplatin. Correlation of CSC expression was performed with known prognostic factors. The effect of stem cell expression on disease outcomes was tested within multivariate analysis. RESULTS: One hundred fifty patients were included. The median dose to point A was 83 Gy (46-89 Gy) and a median of 4 cycles (range, 0-6 cycles) of chemotherapy was administered. At baseline, moderate to strong immunohistochemical expression of SOX-2, OCT-4, Nanog, CD44, and Podoplanin was observed in 12.8%, 4.8%, 24.4%, 15.5%, and 1.3% of patients, respectively. At median follow-up of 30 months (range, 3-51 months), locoregional and distant relapse was observed in 12.2% and 23.1% of patients, of whom 4.7% had both local and distant relapse. The 3-year disease-free survival rate was 87%. On multivariate analysis, moderate to high CSC expression and CD44 low status (hazard ratio [HR] = 8.8; 95% confidence interval [CI], 1.0-77.2; P < .04) independently predicted for locoregional relapse-free survival. International Federation of Gynecology and Obstetrics stage (HR = 2.6; 95% CI, 1.3-5.4; P = .004) and presence of residual tumor after external radiation (HR = 3.5; 95% CI, 1.8-6.5; P = .0001) predicted for a detriment in disease-free survival. CONCLUSIONS: The presence of stem cell proteins and loss of CD44 independently predicts for reduced locoregional control in locally advanced cervical cancer. Further investigation into the interaction of stem cell and CD44 biology is warranted.


Subject(s)
Chemoradiotherapy , Hyaluronan Receptors/analysis , Neoplastic Stem Cells/chemistry , Uterine Cervical Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Octamer Transcription Factor-3/analysis , Prospective Studies , SOXB1 Transcription Factors/analysis , Translational Research, Biomedical , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology
15.
Sci Rep ; 7(1): 11619, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28912581

ABSTRACT

Secretory phospholipase A2 Group-IIA (sPLA2-IIA) is involved in lipid catabolism and growth promoting activity. sPLA2-IIA is deregulated in many pathological conditions including various cancers. Here, we have studied the role of sPLA2-IIA in the development of cyclic alopecia and wound healing response in relation to complete loss of hair follicle stem cells (HFSCs). Our data showed that overexpression of sPLA2-IIA in homozygous mice results in hyperproliferation and terminal epidermal differentiation followed by hair follicle cycle being halted at anagen like stage. In addition, sPLA2-IIA induced hyperproliferation leads to compl pathological conditions including various cancers. Here ete exhaustion of hair follicle stem cell pool at PD28 (Postnatal day). Importantly, sPLA2-IIA overexpression affects the hair shaft differentiation leading to development of cyclic alopecia. Molecular investigation study showed aberrant expression of Sox21, Msx2 and signalling modulators necessary for proper differentiation of inner root sheath (IRS) and hair shaft formation. Further, full-thickness skin wounding on dorsal skin of K14-sPLA2-IIA homozygous mice displayed impaired initial healing response. Our results showed the involvement of sPLA2-IIA in regulation of matrix cells differentiation, hair shaft formation and complete loss of HFSCs mediated impaired wound healing response. These novel functions of sPLA2-IIA may have clinical implications in alopecia, cancer development and ageing.


Subject(s)
Alopecia/etiology , Alopecia/pathology , Group II Phospholipases A2/genetics , Hair Follicle/metabolism , Hair Follicle/pathology , Hair/pathology , Wound Healing/genetics , Alopecia/metabolism , Animals , Biomarkers , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Regulation , Genotype , Group II Phospholipases A2/metabolism , Mice , Mice, Transgenic , Phenotype , Signal Transduction
16.
Stem Cells ; 34(9): 2407-17, 2016 09.
Article in English | MEDLINE | ID: mdl-27299855

ABSTRACT

Secretory phospholipase A2 Group-IIA (sPLA2 -IIA) catalyzes the hydrolysis of the sn-2 position of glycerophospholipids to yield fatty acids and lysophospholipids. sPLA2 -IIA is deregulated in various cancers; however, its role in hair follicle stem cell (HFSC) regulation is obscure. Here we report a transgenic mice overexpressing sPLA2 -IIA (K14-sPLA2 -IIA) showed depletion of HFSC pool. This was accompanied with increased differentiation, loss of ortho-parakeratotic organization and enlargement of sebaceous gland, infundibulum and junctional zone. The colony forming efficiency of keratinocytes was significantly reduced. Microarray profiling of HFSCs revealed enhanced level of epithelial mitogens and transcription factors, c-Jun and FosB that may be involved in proliferation and differentiation. Moreover, K14-sPLA2 -IIA keratinocytes showed enhanced activation of EGFR and JNK1/2 that led to c-Jun activation, which co-related with enhanced differentiation. Further, depletion of stem cells in bulge is associated with high levels of chromatin silencing mark, H3K27me3 and low levels of an activator mark, H3K9ac suggestive of alteration in gene expression contributing toward stem cells differentiation. Our results, first time uncovered that overexpression of sPLA2 -IIA lead to depletion of HFSCs and differentiation associated with altered histone modification. Thus involvement of sPLA2 -IIA in stem cells regulation and disease pathogenesis suggest its prospective clinical implications. Stem Cells 2016;34:2407-2417.


Subject(s)
Cell Differentiation , Epidermis/enzymology , Group II Phospholipases A2/metabolism , Hair Follicle/cytology , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Stem Cells/cytology , Aging/metabolism , Animals , Cell Proliferation , Enzyme Activation , ErbB Receptors/metabolism , Gene Expression Profiling , Histones/metabolism , Homeostasis , Hyperplasia , Keratinocytes/metabolism , Lysine/metabolism , Methylation , Mice, Transgenic , Parakeratosis/pathology , Sebaceous Glands/pathology , Signal Transduction , Stem Cells/metabolism
17.
J Biomol Struct Dyn ; 33(6): 1291-301, 2015.
Article in English | MEDLINE | ID: mdl-25105795

ABSTRACT

ABRAXAS is an integral member of BRCA1-complex, which helps in its recruitment to the DNA damage site. It interacts with BRCA1 via its C-terminal phospho-peptide binding motif while the N-terminal associates with RAP80, and thereby recruits the BRCA1-complex at the site of DNA damage. Nonetheless, how ABRAXAS helps in the structural integrity of BRCA1-complex, and its DNA repair mechanism remains elusive. To elucidate the role of ABRAXAS in the DNA repair process, we characterized the ABRAXAS wild type and Arg361Gln mutant using in silico and in vitro approach. It has been observed that ABRAXAS Arg361Gln mutant is responsible for defective nuclear localization of BRCA1-complex, and hence important for DNA repair function. We found conformational changes in ABRAXAS mutant, which impaired binding to RAP80 and further disturb BRCA1-complex localization. The results presented in this paper will help to understand the cause of BRCA1 mislocalization, and various DNA repair defects that occur due to substitution. Comparative study of ABRAXAS wild type and mutant will provide helpful perspective for inhibitor designing that can potentially recompense the deleterious effect(s) of Arg361Gln mutation, and have therapeutic application.


Subject(s)
BRCA1 Protein/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Mutation , Amino Acid Sequence , Amino Acid Substitution , BRCA1 Protein/chemistry , Carrier Proteins/chemistry , Codon , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Molecular Weight , Protein Binding , Protein Conformation , Protein Stability , Protein Transport , Thermodynamics
18.
PLoS One ; 8(9): e72707, 2013.
Article in English | MEDLINE | ID: mdl-24039796

ABSTRACT

Receptor Associated Protein 80 (RAP80) is a member of RAP80-BRCA1-CCDC98 complex family and helps in its recruitment to the DNA damage site for effective homologous recombination repair. It encompasses two tandem UIMs (UIM1 and UIM2) motif at its N-terminus, which interact with K-63 linked polyubiquitin chain(s) on H2AX and thereby assemble the RAP80-BRCA1 complex at the damage site. Nevertheless, how RAP80 helps in the structural integrity of BRCA1 complex is still elusive. Considering the role of RAP80 in the recruitment of BRCA1 complex at the DNA damage site, we attempted to explore the molecular mechanism associated with RAP80 and mutation that causes chromosomal aberrations due to its loss of function. There is a significant loss in structural characteristics of RAP80 ΔE81, which impairs its binding affinity with the polyubiquitin chain. This leads to the defective recruitment of RAP80 and BRCA1 complex at the DNA damage site. The results presented here are very useful in understanding the cause of various repair defects (chromosomal aberration) that arise due to this mutation. Comparative study of wild type and ΔE81 could be helpful in designing the small molecules that can potentially compensate the deleterious effect(s) of ΔE81 and hence useful for therapeutic application.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Carrier Proteins/genetics , DNA-Binding Proteins , Histone Chaperones , Humans , Models, Molecular , Molecular Sequence Data , Molecular Weight , Mutation , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Recombinant Proteins , Sequence Alignment , Thermodynamics
19.
Chromosome Res ; 21(3): 203-12, 2013 May.
Article in English | MEDLINE | ID: mdl-23681654

ABSTRACT

Tissue stem cells have been proposed to segregate the chromosomes asymmetrically (in a non-random manner), thereby retaining preferentially the older "immortal" DNA strands bearing the stemness characteristics into one daughter cell, whereas the newly synthesized strands are segregated to the other daughter cell that will commit to differentiation. Moreover, this non-random segregation would protect the stem cell genome from accumulating multiple mutations during repeated DNA replication. This long-standing hypothesis remains an active subject of study due to conflicting results for some systems and lack of consistency among different tissue stem cell populations. In this review, we will focus on work done in the hair follicle, which is one of the best-understood vertebrate tissue stem cell system to date. In cell culture analysis of paired cultured keratinocytes derived from hair follicle, stem cells suggested a non-random segregation of chromosome with respect to the older DNA strand. In vivo, the hair follicle stem cells appear to self-renew and differentiate at different phases of their homeostatic cycle. The fate decisions occur in quiescence when some stem cells migrate out of their niche and commit to differentiation without self-renewal. The stem cells left behind in the niche self-renew symmetrically and randomly segregate the chromosomes at each division, making more stem cells. This model seems to apply to at least a few other vertebrate tissue stem cells in vivo.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , DNA/metabolism , Hair Follicle/cytology , Hair Follicle/metabolism , Homeostasis , Animals , Chromosomes/metabolism , Humans , Organ Specificity
20.
J Surg Res ; 171(2): e223-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21962810

ABSTRACT

BACKGROUND: Since there are no pig embryonic stem cells, pig genetic engineering is done in fetal fibroblasts that remain totipotent for only 3 to 5 wk. Nuclear donor cells that remain totipotent for longer periods of time would facilitate complicated genetic engineering in pigs. The goal of this study was to test the feasibility of using fetal liver-derived cells (FLDC) to perform gene targeting, and create a genetic knockout pig. MATERIALS AND METHODS: FLDC were isolated and processed using a human liver stem cell protocol. Single copy α-1,3-galactosyl transferase knockout (GTKO) FLDCs were created using electroporation and neomycin resistant colonies were screened using PCR. Homozygous GTKO cells were created through loss of heterozygosity mutations in single GTKO FLDCs. Double GTKO FLDCs were used in somatic cell nuclear transfer (SCNT) to create GTKO pigs. RESULTS: FLDCs grew for more than 80 population doublings, maintaining normal karyotype. Gene targeting and loss of heterozygosity mutations produced homozygous GTKO FLDCs. FLDCs used in SCNT gave rise to homozygous GTKO pigs. CONCLUSIONS: FDLCs can be used in gene targeting and SCNT to produce genetically modified pigs. The increased life span in culture compared to fetal fibroblasts may facilitate genetic engineering in the pig.


Subject(s)
Animals, Genetically Modified/genetics , Cloning, Organism/methods , Embryonic Stem Cells/cytology , Gene Targeting/methods , Liver/cytology , Swine/genetics , Animals , Cell Culture Techniques/methods , Embryonic Stem Cells/physiology , Feasibility Studies , Female , Fetus/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Gene Knockout Techniques , Genetic Engineering/methods , Homozygote , Loss of Heterozygosity/genetics , Male , Nuclear Transfer Techniques , Pregnancy , Tail/cytology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...