1
|
Ortega MA, Boaru DL, De Leon-Oliva D, De Castro-Martinez P, Minaya-Bravo AM, Casanova-Martín C, Barrena-Blázquez S, Garcia-Montero C, Fraile-Martinez O, Lopez-Gonzalez L, Saez MA, Alvarez-Mon M, Diaz-Pedrero R. The Impact of Klotho in Cancer: From Development and Progression to Therapeutic Potential. Genes (Basel) 2025; 16:128. [PMID: 40004457 PMCID: PMC11854833 DOI: 10.3390/genes16020128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Klotho, initially identified as an anti-aging gene, has been shown to play significant roles in cancer biology. Alongside α-Klotho, the β-Klotho and γ-Klotho isoforms have also been studied; these studies showed that Klotho functions as a potential tumor suppressor in many different cancers by inhibiting cancer cell proliferation, inducing apoptosis and modulating critical signaling pathways such as the Wnt/β-catenin and PI3K/Akt pathways. In cancers such as breast cancer, colorectal cancer, hepatocellular carcinoma, ovarian cancer, and renal cell carcinoma, reduced Klotho expression often correlates with a poor prognosis. In addition, Klotho's role in enhancing chemotherapy sensitivity and its epigenetic regulation further underscores its potential as a target for cancer treatments. This review details Klotho's multifaceted contributions to cancer suppression and its potential as a therapeutic target, enhancing the understanding of its significance in cancer treatment and prognoses.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Ana M. Minaya-Bravo
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Carlos Casanova-Martín
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
| | - Cielo Garcia-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, Central University Hospital of Defence—UAH Madrid, 28801 Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, Network Biomedical Research Center for Liver and Digestive Diseases (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain; (D.L.B.); (D.D.L.-O.); (P.D.C.-M.); (A.M.M.-B.); (S.B.-B.); (C.G.-M.); (O.F.-M.); (M.A.S.); (M.A.-M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (L.L.-G.); (R.D.-P.)
- Department of General and Digestive Surgery, Príncipe de Asturias, University Hospital, 28805 Alcala de Henares, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| |
Collapse
|
2
|
El-Gazar AA, El-Emam SZ, M El-Sayyad S, El-Mancy SS, Fayez SM, Sheta NM, Al-Mokaddem AK, Ragab GM. Pegylated polymeric micelles of boswellic acid-selenium mitigates repetitive mild traumatic brain injury: Regulation of miR-155 and miR-146a/BDNF/ Klotho/Foxo3a cue. Int Immunopharmacol 2024; 134:112118. [PMID: 38705029 DOI: 10.1016/j.intimp.2024.112118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/07/2024] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
This study aims to explore the protective machinery of pegylated polymeric micelles of boswellic acid-selenium (PMBS) against secondary neuronal damage triggered by mild repetitive traumatic brain injury (RTBI). After PMBS characterization in terms of particle size, size distribution, zeta potential, and transmission electronic microscopy, the selected formula was used to investigate its potency against experimental RTBI. Five groups of rats were used; group 1 (control) and the other four groups were subjected to RTBI. Groups 2 was RTBI positive control, while 3, 4, and 5 received boswellic acid (BSA), selenium (SEL), and PMBS, respectively. The open-field behavioral test was used for behavioral assessment. Subsequently, brain tissues were utilized for hematoxylin and eosin staining, Nissl staining, Western blotting, and ELISA in addition to evaluating microRNA expression (miR-155 and miR-146a). The behavioral changes, oxidative stress, and neuroinflammation triggered by RTBI were all improved by PMBS. Moreover, PMBS mitigated excessive glutamate-induced excitotoxicity and the dysregulation in miR-155 and miR-146a expression. Besides, connexin43 (Cx43) expression as well as klotho and brain-derived neurotrophic factor (BDNF) were upregulated with diminished neuronal cell death and apoptosis because of reduced Forkhead Box class O3a(Foxo3a) expression in the PMBS-treated group. The current study has provided evidence of the benefits produced by incorporating BSA and SEL in PEGylated polymeric micelles formula. PMBS is a promising therapy for RTBI. Its beneficial effects are attributed to the manipulation of many pathways, including the regulation of miR-155 and miR-146a expression, as well as the BDNF /Klotho/Foxo3a signaling pathway.
Collapse
Affiliation(s)
- Amira A El-Gazar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza, Egypt.
| | - Soad Z El-Emam
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Shorouk M El-Sayyad
- Pharmacology and Toxicology Department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Shereen S El-Mancy
- Pharmaceutics and industrial pharmacy department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Sahar M Fayez
- Pharmaceutics and industrial pharmacy department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Nermin M Sheta
- Pharmaceutics and industrial pharmacy department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ghada M Ragab
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12585, Egypt
| |
Collapse
|
3
|
Zhou M, Niu H, Chen R, Chen W, Cui D. Gui ShenWan prevent premature ovarian insufficiency by modulating autophagy and angiogenesis via facilitating VDR. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117955. [PMID: 38395181 DOI: 10.1016/j.jep.2024.117955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 02/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gui Shen Wan (GSW) stands out as a promising therapeutic approach for addressing Premature Ovarian Insufficiency (POI). With deep roots in traditional medicine, GSW highlights the ethnopharmacological significance of herbal interventions in addressing nuanced aspects of women's health, with a specific emphasis on ovarian functionality. Recognizing the importance of GSW in gynecological contexts resonates with a rich tradition of using botanical formulations to navigate the intricacies of reproductive health. Delving into GSW's potential for treating POI emphasizes the crucial role of ethnopharmacological insights in guiding modern research endeavors. AIM OF THE STUDY GSW is extensively utilized in gynecological disorders and has recently emerged as a potential therapeutic approach for POI. The present investigation aimed to assess the efficacy of GSW in treating POI in rats and elucidate its underlying molecular mechanisms. MATERIALS AND METHODS The study employed GSW for POI treatment in rats. GSW, prepared as pills, underwent HPLC fingerprinting for quality control. Reagents and drugs, including VCD and dehydroepiandrosterone (DHEA), were sourced from reputable providers. Eighty Sprague-Dawley rats were categorized into groups for POI induction and treatment. Ovarian tissue underwent HE staining, immunohistochemical staining, Western Blot, qRT-PCR, and vaginal secretion testing. ELISA was utilized for target molecule detection. This methodology ensures a robust and reliable experimental framework. RESULTS The results highlight a robust collaborative improvement in POI among rats subjected to combined GSW and DHEA treatment. Particularly noteworthy is the substantial enhancement in the expression of vascular regeneration-related molecules-VDR-Klotho-VEGFR-accompanied by a significant elevation in autophagy levels. Post-GSW administration, rat ovarian morphology demonstrated increased stability, hormone levels exhibited more consistent maintenance, and there was a marked reduction in inflammatory response compared to other groups (p < 0.01). Furthermore, GSW intervention resulted in a more pronounced upregulation of ovarian autophagy (p < 0.05). CONCLUSION By modulating VDR-Klotho signaling, GSW exerts regulatory control over ovarian autophagy and vascular regeneration, thereby mitigating the occurrence and progression of POI in rats.
Collapse
Affiliation(s)
- Minfeng Zhou
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China
| | - Huifang Niu
- School of Food Science and Technology, Huazhong Agricultural University, 1 Lion Rock Street. Wuhan City, 430070, China
| | - Rui Chen
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China.
| | - Wenmao Chen
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China.
| | - Dandan Cui
- Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, 1277 Liberation Avenue, 430022, Wuhan City, China.
| |
Collapse
|
4
|
Soykan MN, Gunes S. Overexpression of Klotho gene using CRISPR/Cas9 induces apoptosis and inhibits cell motility in the human colorectal cancer cells. Biotechnol J 2024; 19:e2300496. [PMID: 38403402 DOI: 10.1002/biot.202300496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 02/27/2024]
Abstract
Despite advances in early detection and treatment, colorectal cancer remains one of the leading causes of cancer-related deaths. The klotho (KL) gene plays a critical role in the development and progression of colorectal cancer. This study investigates the role of the KL gene in colorectal cancer by using the CRISPR/Cas9 system to overexpress and knock out (KO) the KL gene in human colorectal cancer cells (Caco-2). The effects of the changes were assessed by gene expression analysis, flow cytometry, scratch wound closure assays, colony formation assays, and immunofluorescence staining. Our results showed that overexpression of the KL gene increased apoptosis and decreased cell motility in cancer cells, whereas knockout of the KL gene had the opposite role. The present study elucidates the mechanisms underlying this role and highlights the potential of the CRISPR/Cas9 system as a gene editing tool in cancer research. Our data suggest that activation of the KL gene may serve as a novel therapeutic strategy and biomarker for studies in colorectal cancer.
Collapse
Affiliation(s)
- Merve Nur Soykan
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Sibel Gunes
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkey
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
5
|
Mota J, Lima AMM, Gomes JIS, Souza de Andrade M, Brito HO, Silva MMAL, Faustino-Rocha AI, Oliveira PA, Lopes FF, Gil da Costa RM. Klotho in Cancer: Potential Diagnostic and Prognostic Applications. Diagnostics (Basel) 2023; 13:3357. [PMID: 37958253 PMCID: PMC10650206 DOI: 10.3390/diagnostics13213357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Klotho proteins, αKlotho, βKlotho, and γKlotho, exert tumor-suppressive activities via the fibroblast growth factor receptors and multiple cell-signaling pathways. There is a growing interest in Klotho proteins as potential diagnostic and prognostic biomarkers for multiple diseases. However, recent advances regarding their roles and potential applications in cancer remain disperse and require an integrated analysis. The present review analyzed research articles published between 2012 and 2022 in the Cochrane and Scopus scientific databases to study the role of Klotho in cancer and their potential as tools for diagnosing specific cancer types, predicting tumor aggressiveness and prognosis. Twenty-six articles were selected, dealing with acute myeloid leukemia and with bladder, breast, colorectal, esophageal, gastric, hepatocellular, ovarian, pancreatic, prostatic, pulmonary, renal, and thyroid cancers. αKlotho was consistently associated with improved prognosis and may be useful in estimating patient survival. A single study reported the use of soluble αKlotho levels in blood serum as a tool to aid the diagnosis of esophageal cancer. γKlotho was associated with increased aggressiveness of bladder, breast, and prostate cancer, and βKlotho showed mixed results. Further clinical development of Klotho-based assays will require careful identification of specific tumor subtypes where Klotho proteins may be most valuable as diagnostic or prognostic tools.
Collapse
Affiliation(s)
- Jucileide Mota
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Alice Marques Moreira Lima
- Health Sciences Center, State University of the Tocantins Region of Maranhão (UEMASUL), Imperatriz 6591-480, Brazil
| | - Jhessica I. S. Gomes
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Marcelo Souza de Andrade
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Haissa O. Brito
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
- Morphology Department, Federal University of Maranhão, São Luís 65085-580, Brazil
| | | | - Ana I. Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Fernanda F. Lopes
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
| | - Rui M. Gil da Costa
- Post-Graduate Programme in Adult Health (PPGSAD), Federal University of Maranhão, São Luís 65085-580, Brazil
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Inov4Agro—Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, 4200-465 Porto, Portugal
- Associate Laboratory in Chemical Engineering, Faculty of Engineering (ALiCE), University of Porto, 4200-465 Porto, Portugal
- Molecular Oncology and Viral Pathology Group, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
- Health Research Network, Research Center of Portuguese Oncology Institute of Porto (CIIPOP/RISE@CIIPOP), 4200-072 Porto, Portugal
| |
Collapse
|
6
|
Czegle I, Huang C, Soria PG, Purkiss DW, Shields A, Wappler-Guzzetta EA. The Role of Genetic Mutations in Mitochondrial-Driven Cancer Growth in Selected Tumors: Breast and Gynecological Malignancies. Life (Basel) 2023; 13:996. [PMID: 37109525 PMCID: PMC10145875 DOI: 10.3390/life13040996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
There is an increasing understanding of the molecular and cytogenetic background of various tumors that helps us better conceptualize the pathogenesis of specific diseases. Additionally, in many cases, these molecular and cytogenetic alterations have diagnostic, prognostic, and/or therapeutic applications that are heavily used in clinical practice. Given that there is always room for improvement in cancer treatments and in cancer patient management, it is important to discover new therapeutic targets for affected individuals. In this review, we discuss mitochondrial changes in breast and gynecological (endometrial and ovarian) cancers. In addition, we review how the frequently altered genes in these diseases (BRCA1/2, HER2, PTEN, PIK3CA, CTNNB1, RAS, CTNNB1, FGFR, TP53, ARID1A, and TERT) affect the mitochondria, highlighting the possible associated individual therapeutic targets. With this approach, drugs targeting mitochondrial glucose or fatty acid metabolism, reactive oxygen species production, mitochondrial biogenesis, mtDNA transcription, mitophagy, or cell death pathways could provide further tailored treatment.
Collapse
Affiliation(s)
- Ibolya Czegle
- Department of Internal Medicine and Haematology, Semmelweis University, H-1085 Budapest, Hungary
| | - Chelsea Huang
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Priscilla Geraldine Soria
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Dylan Wesley Purkiss
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Andrea Shields
- Department of Pathology and Laboratory Medicine, Loma Linda University Health, Loma Linda, CA 92354, USA
| | | |
Collapse
|
7
|
The role of α-klotho in human cancer: molecular and clinical aspects. Oncogene 2022; 41:4487-4497. [PMID: 36038662 DOI: 10.1038/s41388-022-02440-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/14/2022] [Accepted: 08/10/2022] [Indexed: 11/08/2022]
Abstract
Klotho is a well-established longevity hormone. Its most prominent function is the regulation of phosphate homeostasis. However, klotho possesses multiple pleiotropic activities, including inhibition of major signaling pathways, reducing oxidative stress and suppressing inflammation. These activities are tightly associated with cancer, and klotho was discovered as a universal tumor suppressor. We review here novel molecular aspects of klotho activity in cancer, focusing on its structure-function relationships and clinical aspects regarding its expression, blood levels, clinical risk, and prognostic value in the clinical setting. In addition, the potential benefit of klotho treatment combined with chemotherapy, biological therapy, or immunotherapy, are discussed. Finally, as klotho was shown in preclinical models to inhibit cancer development and growth, we discuss various approaches to developing klotho-based therapies.
Collapse
|
8
|
Li C, Jiang S, Wang H, Wang Y, Han Y, Jiang J. Berberine exerts protective effects on cardiac senescence by regulating the Klotho/SIRT1 signaling pathway. Biomed Pharmacother 2022; 151:113097. [PMID: 35609366 DOI: 10.1016/j.biopha.2022.113097] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/04/2022] [Accepted: 05/07/2022] [Indexed: 11/15/2022] Open
Abstract
Berberine (BBR), an isoquinoline alkaloid, exerts protective effects on various cardiac injuries, and also extends the lifespan of individuals. However, the cardioprotective effect of BBR on cardiac senescence remains unknown. This study investigated the effects of BBR on cardiac senescence and its underlying mechanism. Senescent H9c2 cells induced by doxorubicin (DOX) and naturally aged rats were used to evaluate the protective effects of BBR on cardiac senescence. The results showed that BBR protected H9c2 cells against DOX-induced senescence. Exogenous Klotho (KL) exerts similar effects to those of BBR. BBR significantly increased in protein expression of KL, while transfection with KL-specific siRNA (siKL) inhibited the protective effect of BBR against senescence. Both BBR and exogenous KL decreased the levels of reactive oxygen species, inhibited apoptosis, and alleviated mitochondrial dysfunction in these cells; and transfection with siKL attenuated these effects of BBR. In naturally aged rats, BBR indeed protected the animals from cardiac aging, at least partially, through lowering the levels of cardiac hypertrophy markers, and increased the expression of KL in cardiac tissue. Additionally, BBR markedly reversed downregulation of sirtuin1 (SIRTI) in the aged heart. In vitro experiments revealed that BBR and exogenous KL also increased the expression of SIRT1, whereas siKL limited this effect of BBR in senescent H9c2 cell. In summary, BBR upregulated KL expression and prevented heart from cardiac senescence through anti-oxidative and anti-apoptotic effects, as well as alleviation of mitochondrial dysfunction. These effects may be mediated via regulation of the Klotho/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Cong Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Hengfei Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, 100050 Beijing, China.
| |
Collapse
|
9
|
Borges BC, Do Amaral PA, Soldi LR, Costa Silva VL, Carvalho De Souza F, Cordeiro Da Luz FA, Agenor De Araújo R, Barbosa Silva MJ. Undetected αKlotho in serum is associated with the most aggressive phenotype of breast cancer. Mol Clin Oncol 2022; 16:93. [PMID: 35350405 PMCID: PMC8943645 DOI: 10.3892/mco.2022.2526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/09/2021] [Indexed: 11/06/2022] Open
Abstract
Klotho, a cellular anti-senescence protein, is related to antitumor actions, growth regulation, proliferation and invasiveness in several types of tumor, including breast cancer. The present study aimed to analyze the serum levels of αKlotho in patients with breast cancer according to histopathological and immunohistochemical variables. A total of 74 patients and 60 healthy controls were recruited. Peripheral blood samples were collected and serum levels were assessed by sandwich ELISA. Clinical and diagnostic data were obtained from medical records and databases of the Clinical Hospital of the Federal University of Uberlândia (Uberlândia, Brazil). The results indicated no difference in the levels of αKlotho between patients and controls (P=0.068); however, the number of patients with breast cancer with undetectable αKlotho was high (n=52). Thus, the variables that were associated with the lowest survival rates were analyzed, relating them to undetectable αKlotho. Among cases of metastatic tumors or tumors with poor differentiation, positive lymph node status and triple-negative status, patients with undetectable αKlotho predominated and had unfavorable overall survival. Due to the significant results obtained in triple-negative patients, an in vitro analysis was performed to determine whether estrogen receptors (ERs) have a role in αKlotho production. Treatment of MCF-7 cells with ER agonists, estradiol (E2) and diarylpropionitrile (DPN), resulted in increases in αKlotho expression and supernatant levels of both agonists, demonstrating a direct association between the ER and Klotho production; of note, the ERβ-specific agonist DPN tripled αKlotho expression when compared to E2 (P=0.078). These data suggested that undetectable αKlotho in the serum of patients with breast cancer is related to unfavorable histopathological variables and poor prognosis and ERs possibly have an important role in maintaining adequate quantities of αKlotho.
Collapse
Affiliation(s)
- Bruna Cristina Borges
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| | - Pedro Augusto Do Amaral
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| | - Luiz Ricardo Soldi
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| | - Victor Luigi Costa Silva
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| | - Fernanda Carvalho De Souza
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| | - Felipe Andrés Cordeiro Da Luz
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
- Núcleo de Prevenção, Pesquisa e Projeto de Câncer, Hospital do Câncer, Uberlândia, MG 38400-902, Brazil
| | - Rogério Agenor De Araújo
- Núcleo de Prevenção, Pesquisa e Projeto de Câncer, Hospital do Câncer, Uberlândia, MG 38400-902, Brazil
| | - Marcelo José Barbosa Silva
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG 38400-902, Brazil
| |
Collapse
|
10
|
Arbel Rubinstein T, Reuveni I, Hesin A, Klein-Goldberg A, Olauson H, Larsson TE, Abraham CR, Zeldich E, Bosch A, Chillón M, Hollander KS, Shabtay-Orbach A, Vainer GW, Wolf I, Rubinek T. A Transgenic Model Reveals the Role of Klotho in Pancreatic Cancer Development and Paves the Way for New Klotho-Based Therapy. Cancers (Basel) 2021; 13:cancers13246297. [PMID: 34944918 PMCID: PMC8699737 DOI: 10.3390/cancers13246297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary We aimed to study the role of the anti-aging protein klotho and its secreted isoform, sKL, in pancreatic cancer. Three in vivo models, including a novel genetic mouse model and bioinformatics analyses, indicated klotho as a tumor suppressor in pancreatic ductal adenocarcinoma, and unveiled a unique klotho DNA hypermethylation pattern in pancreatic tumors. These results possess significant prognostic value, and further suggest that sKL may serve as a therapeutic agent for pancreatic ductal adenocarcinoma. Abstract Klotho is an anti-aging transmembrane protein, which can be shed and can function as a hormone. Accumulating data indicate that klotho is a tumor suppressor in a wide array of malignancies, and designate the subdomain KL1 as the active region of the protein towards this activity. We aimed to study the role of klotho as a tumor suppressor in pancreatic ductal adenocarcinoma (PDAC). Bioinformatics analyses of The Cancer Genome Atlas (TCGA) datasets revealed a correlation between the survival of PDAC patients, levels of klotho expression, and DNA methylation, and demonstrated a unique hypermethylation pattern of klotho in pancreatic tumors. The in vivo effects of klotho and KL1 were examined using three mouse models. Employing a novel genetic model, combining pancreatic klotho knockdown with a mutation in Kras, the lack of klotho contributed to PDAC generation and decreased mousece survival. In a xenograft model, administration of viral particles carrying sKL, a spliced klotho isoform containing the KL1 domain, inhibited pancreatic tumors. Lastly, treatment with soluble sKL prolonged survival of Pdx1-Cre; KrasG12D/+;Trp53R172H/+ (KPC) mice, a model known to recapitulate human PDAC. In conclusion, this study provides evidence that klotho is a tumor suppressor in PDAC. Furthermore, these data suggest that the levels of klotho expression and DNA methylation could have prognostic value in PDAC patients, and that administration of exogenous sKL may serve as a novel therapeutic strategy to treat PDAC.
Collapse
Affiliation(s)
- Tammi Arbel Rubinstein
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Inbal Reuveni
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Arkadi Hesin
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anat Klein-Goldberg
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden; (H.O.); (T.E.L.)
| | - Tobias E. Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 14186 Stockholm, Sweden; (H.O.); (T.E.L.)
- Department of Nephrology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Carmela R. Abraham
- Boston University School of Medicine, Department of Biochemistry, Boston, MA 02118, USA; (C.R.A.); (E.Z.)
- Pharmacology & Experimental Therapeutics, Boston, MA 02118, USA
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
| | - Ella Zeldich
- Boston University School of Medicine, Department of Biochemistry, Boston, MA 02118, USA; (C.R.A.); (E.Z.)
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
| | - Assumpció Bosch
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
- Institut de Neurociències, Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
- CIBERNED, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Chillón
- Klogenix Therapeutics Inc., Boston, MA 02116, USA; (A.B.); (M.C.)
- Institut de Neurociències, Department of Biochemistry and Molecular Biology, Universitat Autonoma de Barcelona, 08035 Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall d’Hebron Institut de Recerca (VHIR), 08035 Barcelona, Spain
- ICREA, Institut Catalan Recerca Avançada, 08010 Barcelona, Spain
| | - Kenneth Samuel Hollander
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Ayelet Shabtay-Orbach
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
| | - Gilad W. Vainer
- Pathology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
| | - Ido Wolf
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tami Rubinek
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv 64239, Israel; (T.A.R.); (I.R.); (A.H.); (A.K.-G.); (K.S.H.); (A.S.-O.); (I.W.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-527466151
| |
Collapse
|
11
|
Ewendt F, Feger M, Föller M. Role of Fibroblast Growth Factor 23 (FGF23) and αKlotho in Cancer. Front Cell Dev Biol 2021; 8:601006. [PMID: 33520985 PMCID: PMC7841205 DOI: 10.3389/fcell.2020.601006] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/15/2020] [Indexed: 12/16/2022] Open
Abstract
Together with fibroblast growth factors (FGFs) 19 and 21, FGF23 is an endocrine member of the family of FGFs. Mainly secreted by bone cells, FGF23 acts as a hormone on the kidney, stimulating phosphate excretion and suppressing formation of 1,25(OH)2D3, active vitamin D. These effects are dependent on transmembrane protein αKlotho, which enhances the binding affinity of FGF23 for FGF receptors (FGFR). Locally produced FGF23 in other tissues including liver or heart exerts further paracrine effects without involvement of αKlotho. Soluble Klotho (sKL) is an endocrine factor that is cleaved off of transmembrane Klotho or generated by alternative splicing and regulates membrane channels, transporters, and intracellular signaling including insulin growth factor 1 (IGF-1) and Wnt pathways, signaling cascades highly relevant for tumor progression. In mice, lack of FGF23 or αKlotho results in derangement of phosphate metabolism and a syndrome of rapid aging with abnormalities affecting most organs and a very short life span. Conversely, overexpression of anti-aging factor αKlotho results in a profound elongation of life span. Accumulating evidence suggests a major role of αKlotho as a tumor suppressor, at least in part by inhibiting IGF-1 and Wnt/β-catenin signaling. Hence, in many malignancies, higher αKlotho expression or activity is associated with a more favorable outcome. Moreover, also FGF23 and phosphate have been revealed to be factors relevant in cancer. FGF23 is particularly significant for those forms of cancer primarily affecting bone (e.g., multiple myeloma) or characterized by bone metastasis. This review summarizes the current knowledge of the significance of FGF23 and αKlotho for tumor cell signaling, biology, and clinically relevant parameters in different forms of cancer.
Collapse
Affiliation(s)
- Franz Ewendt
- Department of Nutritional Physiology, Institute of Agricultural and Nutritional Sciences, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
12
|
Klotho rewires cellular metabolism of breast cancer cells through alteration of calcium shuttling and mitochondrial activity. Oncogene 2020; 39:4636-4649. [PMID: 32398866 DOI: 10.1038/s41388-020-1313-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Klotho is a transmembrane protein, which can be shed and act as a circulating hormone and is involved in regulating cellular calcium levels and inhibition of the PI3K/AKT pathway. As a longevity hormone, it protects normal cells from oxidative stress, and as a tumor suppressor it inhibits growth of cancer cells. Mechanisms governing these differential activities have not been addressed. Altered cellular metabolism is a hallmark of cancer and dysregulation of mitochondrial activity is a hallmark of aging. We hypothesized that klotho exerts its differential effects through regulation of these two hallmarks. Treatment with klotho inhibited glycolysis, reduced mitochondrial activity and membrane potential only in cancer cells. Accordingly, global metabolic screen revealed that klotho altered pivotal metabolic pathways, amongst them glycolysis and tricarboxylic acid cycle in breast cancer cells. Alteration of metabolic activity and increased AMP/ATP ratio lead to LKB1-dependent AMPK activation. Indeed, klotho induced AMPK phosphorylation; furthermore, inhibition of LKB1 partially abolished klotho's tumor suppressor activity. By diminishing deltapsi (Δψ) klotho also inhibited mitochondria Ca2+ shuttling thereby impairing mitochondria communication with SOCE leading to reduced Ca2+ influx by SOCE channels. The reduced SOCE was followed by ER Ca2+ depletion and stress. These data delineate mechanisms mediating the differential effects of klotho toward cancer versus normal cells, and indicate klotho as a potent regulator of metabolic activity.
Collapse
|
13
|
Chen B, Huang S, Pisanic Ii TR, Stark A, Tao Y, Cheng B, Li Y, Wei Y, Zhao W, Wang TH, Wu J. Rab8 GTPase regulates Klotho-mediated inhibition of cell growth and progression by directly modulating its surface expression in human non-small cell lung cancer. EBioMedicine 2019; 49:118-132. [PMID: 31707148 PMCID: PMC6945242 DOI: 10.1016/j.ebiom.2019.10.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The klotho (KL) gene is an anti-aging gene that has recently been shown to also function as a general tumor suppressor. However, there is currently only limited information regarding the potential molecular signals for regulation of Klotho without identifying precise molecular mechanisms or interactions. METHODS We performed a mass spectrometry (MS) assay to screen candidate proteins complexed with Klotho derived from immunoprecipitation in human non-small cell lung cancer (NSCLC) cells, and identified Rab8 to be the protein that most prominently interacts with Klotho. We further investigated whether Rab8 can regulate trafficking of Klotho and which process it would modulate using surface biotinylation assay, immunofluorescence and fluorescence ratio microscopy. Furthermore, we explored whether Rab8 is involved in Klotho-mediated function in NSCLC, and verified the results which we found in vivo using xenograft mouse model. FINDINGS We report discovery of Rab8 as a Klotho-interacting protein that acts as a critical modulator of Klotho surface expression in human NSCLC. In particular, we report that Rab8 is co-localized and associated with Klotho, and Klotho trafficking is regulated by Rab8. Moreover, we found that Rab8 modulates surface levels of Klotho via a post-biosynthetic pathway, as opposed to an endocytic pathway. Furthermore, we demonstrate that Rab8 is involved in Klotho-mediated regulation of cell proliferation, migration, invasiveness, epithelial-mesenchymal transition (EMT), and Wnt-β-catenin signaling in NSCLC. Additionally, Rab8 overexpression was also found to increase Klotho-mediated inhibition of NSCLC tumorigenesis in vivo. INTERPRETATION Overall, our findings suggest that Rab8 GTPase can regulate Klotho-mediated inhibition of Wnt signaling activity by modulating translocation of Klotho onto the cell surface, which in turn affects Klotho-mediated inhibition of cell proliferation, migration and invasiveness in NSCLC. These results have important implications for the development of new therapeutic targets, Klotho-related research in the context of NSCLC as well as other areas, and provide a working model for Rab8 function in the context of cancer and cancer biology.
Collapse
Affiliation(s)
- Bo Chen
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Jiangsu Province Hospital, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China; Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Shuhong Huang
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China
| | - Thomas R Pisanic Ii
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Alejandro Stark
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Yong Tao
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States
| | - Bei Cheng
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Yue Li
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Jiangsu Province Hospital, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Yunyan Wei
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Weihong Zhao
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Jiangsu Province Hospital, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China
| | - Tza-Huei Wang
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, United States; Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Jianqing Wu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, Jiangsu Province Hospital, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
14
|
Li F, Li X, Li Z, Ji W, Lu S, Xia W. βKlotho is identified as a target for theranostics in non-small cell lung cancer. Theranostics 2019; 9:7474-7489. [PMID: 31695781 PMCID: PMC6831461 DOI: 10.7150/thno.35582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/11/2019] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a great challenge, calling for the identification of novel molecular targets with diagnostic/therapeutic value. Here, we sought to characterize the expression of βKlotho and its anti-tumor roles in NSCLC. Methods: The expression of βKlotho was examined in NSCLC cells and tissues by western blot, qRT-PCR and immunohistochemistry staining respectively. Biological roles of βKlotho were revealed by a series of functional in vitro and in vivo studies. Serum βKlotho concentrations of patients were measured using specific ELISA methods. Results: Serum βKlotho concentrations of NSCLC patients were significantly lower than the control group. Moreover, βKlotho expression was negatively associated with lymph node metastasis, overall survival and progression-free survival. Overexpression of βKlotho or exogenous βKlotho administration inhibited the proliferation and migration of NSCLC cells, accompanied by induction of apoptosis, G1 to S phase arrest, and inactivation of ERK1/2, AKT and STAT3 signaling. Furthermore, βKlotho overexpression inhibited NSCLC tumor growth in vivo. Conclusions: βKlotho serves as a novel target for theranostics in NSCLC, which has potential clinical applications in the future.
Collapse
Affiliation(s)
- Fan Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiyao Li
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxiang Ji
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Liu Y, Pan J, Pan X, Wu L, Bian J, Lin Z, Xue M, Su T, Lai S, Chen F, Ge Q, Chen L, Ye S, Zhu Y, Chen S, Wang L. Klotho-mediated targeting of CCL2 suppresses the induction of colorectal cancer progression by stromal cell senescent microenvironments. Mol Oncol 2019; 13:2460-2475. [PMID: 31545552 PMCID: PMC6822285 DOI: 10.1002/1878-0261.12577] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 12/23/2022] Open
Abstract
Senescent microenvironments play an important role in tumor progression. Here, we report that doxorubicin (DOX)‐pretreated or replicative senescent stromal cells (WI‐38 and HUVEC) promote colorectal cancer (CRC) cell growth and invasion in vitro and in vivo. These pro‐tumorigenic effects were attenuated by exogenous administration of Klotho, an anti‐aging factor. We subsequently identified several senescence‐associated secretory phenotype (SASP)‐associated genes, including CCL2, which were significantly upregulated in both types of senescent stromal cells during replication and DNA damage‐induced senescence. Importantly, we found that the secretion of CCL2 by senescent stromal cells was significantly higher than that seen in nonsenescent cells or in senescent cells pretreated with Klotho. Notably, CCL2 was found to accelerate CRC cell proliferation and invasion, while this effect could be blocked by administration of a specific CCR2 antagonist. We further show that Klotho can suppress NF‐κB activation during DOX‐induced senescence and thus block CCL2 transcription. Low expression of Klotho, or high expression of CCL2 in patient tumor tissues, correlated with poor overall survival of CRC patients. Collectively, our findings suggest that senescent stromal cells are linked to progression of CRC. Klotho can suppress the senescent stromal cell‐associated triggering of CRC progression by inhibiting the expression of SASP factors including CCL2. The identification of key SASP factors such as CCL2 may provide potential therapeutic targets for improving CRC therapy.
Collapse
Affiliation(s)
- Yangyang Liu
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China.,Department of Gastroenterology, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, China
| | - Jie Pan
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China.,Department of Endocrinology and Metabolism, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Pan
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Lunpo Wu
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Jun Bian
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Zhenghua Lin
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Meng Xue
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Tingting Su
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Sanchuan Lai
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Fei Chen
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Qiwei Ge
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Luyi Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Shufang Ye
- Department of Gastroenterology, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, China
| | - Yabi Zhu
- Department of Gastroenterology, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People's Hospital, China
| | - Shujie Chen
- Institution of Gastroenterology, Zhejiang University, Hangzhou, China.,Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Liangjing Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Institution of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Abolghasemi M, Yousefi T, Maniati M, Qujeq D. The interplay of Klotho with signaling pathway and microRNAs in cancers. J Cell Biochem 2019; 120:14306-14317. [DOI: 10.1002/jcb.29022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Maryam Abolghasemi
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
- Student Research Committee Babol University of Medical Sciences Babol Iran
| | - Tooba Yousefi
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
- Student Research Committee Babol University of Medical Sciences Babol Iran
| | - Mahmood Maniati
- Assistant Professor of the English Department Ahvaz Jundishapur University of Medical Sciences Ahvaz Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
- Department of Clinical Biochemistry, School of Medicine Babol University of Medical Sciences Babol Iran
| |
Collapse
|
17
|
Klotho suppresses colorectal cancer through modulation of the unfolded protein response. Oncogene 2018; 38:794-807. [PMID: 30232408 DOI: 10.1038/s41388-018-0489-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022]
Abstract
Klotho is an anti-aging transmembrane protein, which can be shed and function as a hormone. Accumulating data indicate klotho as a tumor suppressor in a wide array of malignancies and indicate the subdomain KL1 as the active region of the protein. We aimed to study the role of klotho as a tumor suppressor in colorectal cancer. Bioinformatics analyses of TCGA datasets indicated reduced klotho mRNA levels in human colorectal cancer, along with negative regulation of klotho expression by hypermethylation of the promoter and 1st exon, and hypomethylation of an area within the gene. Overexpression or treatment with klotho or KL1 inhibited proliferation of colorectal cancer cells in vitro. The in vivo activity of klotho and KL1 was examined using two models recapitulating development of tumors in the normal colonic environment of immune-competent mice. Treatment with klotho inhibited formation of colon polyps induced by the carcinogen azoxymethane, and KL1 treatment slowed growth of orthotopically-implanted colorectal tumors. Gene expression array revealed that klotho and KL1 expression enhanced the unfolded protein response (UPR) and this was further established by increased levels of spliced XBP1, GRP78 and phosphorylated-eIF2α. Furthermore, attenuation of the UPR partially abrogated klotho tumor suppressor activity. In conclusion, this study indicates klotho as a tumor suppressor in colorectal cancer and identifies, for the first time, the UPR as a pathway mediating klotho activities in cancer. These data suggest that administration of exogenous klotho or KL1 may serve as a novel strategy for prevention and treatment of colorectal cancer.
Collapse
|
18
|
Li Q, Li Y, Liang L, Li J, Luo D, Liu Q, Cai S, Li X. Klotho negatively regulated aerobic glycolysis in colorectal cancer via ERK/HIF1α axis. Cell Commun Signal 2018; 16:26. [PMID: 29884183 PMCID: PMC5994118 DOI: 10.1186/s12964-018-0241-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/28/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Klotho (KL) was originally characterized as an aging suppressor gene, and has been identified as a tumor suppressor gene in a variety of cancers, including colorectal cancer. Recent years have witnessed the importance of metabolism transformation in cancer cell malignancies maintenance. Aberrant cancer cell metabolism is considered to be the hallmark of cancer. Our previous studies demonstrated that KL played negative roles in colon cancer cell proliferation and metastasis. However, its role in the cancer cell reprogramming has seldom been reported. The aim of this study was to examine the role of KL in aerobic glycolysis in colorectal cancer. METHODS Combining maximum standardized uptake value (SUVmax), which was obtained preoperatively via a PET/CT scan, with immunohistochemistry staining, we analyzed the correlation between SUVmax and KL expression in colorectal cancer tissues. The impact of KL on glucose metabolism and its mechanisms were further validated in vitro and in vivo. RESULTS Patients with lower KL expression exhibited higher 18F-FDG uptake (P < 0.05), indicating that KL might participate in aerobic glycolysis regulation. In vitro assay by using colon cancer cell lines further supported this observation. By overexpressing KL in HTC116 and SW480 cells, we observed that the glycolysis was inhibited and the mitochondrial respiration increased, indicating that KL was a negative regulator of aerobic glycolysis. To seek for the underlying mechanisms, we tried to dig out the relation between KL and HIF1α signaling pathway, and found that KL negatively regulated HIF1α protein level and transcriptional activity. Western blot analysis showed that KL overexpression negatively regulated ERK pathway, and KL regulated aerobic glycolysis in part through its regulation of ERK/ HIF1α axis. CONCLUSIONS Taken together, KL is a negative regulator of aerobic glycolysis and KL inhibited glucose metabolism transformation via the ERK/ HIF1α axis.
Collapse
Affiliation(s)
- Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Yaqi Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Jing Li
- Departments of CyberKnife, Huashan Hospital, Fudan University, Shanghai, 200032 China
| | - Dakui Luo
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Qi Liu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
- Department of Oncology, Shanghai Medical College, Fudan University, No.270 Dong’an Road, Xuhui District, Shanghai, 200032 China
| |
Collapse
|
19
|
Dehghani M, Brobey RK, Wang Y, Souza G, Amato RJ, Rosenblatt KP. Klotho inhibits EGF-induced cell migration in Caki-1 cells through inactivation of EGFR and p38 MAPK signaling pathways. Oncotarget 2018; 9:26737-26750. [PMID: 29928482 PMCID: PMC6003560 DOI: 10.18632/oncotarget.25481] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023] Open
Abstract
Klotho is a single-pass transmembrane protein with documented anti-cancer properties. Recent reports have implicated Klotho as an inhibitor of transforming growth factor β1 induced cell migration in renal fibrosis. Overexpression of epidermal growth factor receptor (EGFR) is known to promote tumor initiation and progression in clear-cell renal cell carcinoma (cRCC). We tested our hypothesis that Klotho inhibits EGF-mediated cell migration in cRCC by interfering with the EGFR signaling complex and mitogen-activated protein kinase (MAPK) pathways. We performed cell adhesion, migration, and biochemical studies in vitro using Caki-1 cell line. In addition, we validated the cell culture studies with expression analysis of six de-identified FFPE tissues from primary and metastatic cRCC patients. Our studies show that Klotho inhibited EGF-induced Caki-1 de-adhesion and decreased spreading on collagen type 1. Klotho also inhibited EGF-induced α2β1 integrin-dependent cell migration on collagen type 1. To test the involvement of MAPK pathways in EGF-induced Caki-1 cell motility, the cells were pretreated with either SB203580, a specific p38 MAPK inhibitor, or Klotho. SB203580 blocked the EGF-induced Caki-1 cell migration. Klotho had a comparable inhibitory effect. Our FFPE clinical specimens revealed decreased Klotho mRNA expression compared to a control, non-cancer kidney tissue. The decrease in Klotho mRNA levels correlated with increased c-Src expression, while E-Cadherin was relatively reduced in metastatic FFPE specimens where Klotho was least expressed. Taken together, these results suggest that secreted Klotho inhibits EGF-induced pro-migratory cell morphological changes and migration in Caki-1 cells. Our data additionally suggest that decreased Klotho expression may be involved in cRCC metastasis.
Collapse
Affiliation(s)
- Mehdi Dehghani
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America
| | - Reynolds K Brobey
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America
| | - Yue Wang
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America
| | - Glauco Souza
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America.,n3D Biosciences, Inc., Houston, Texas 77030, United States of America
| | - Robert J Amato
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America
| | - Kevin P Rosenblatt
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas 77030, United States of America.,NX Prenatal, Inc., Bellaire, Texas 77401, United States of America.,Consultative Genomics, PLLC, Bellaire, Texas 77401, United States of America
| |
Collapse
|
20
|
Mao S, Wang X, Wu L, Zang D, Shi W. Association between klotho expression and malignancies risk and progression: A meta-analysis. Clin Chim Acta 2018; 484:14-20. [PMID: 29775618 DOI: 10.1016/j.cca.2018.05.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/30/2018] [Accepted: 05/15/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND We assessed the association between tissue klotho protein expression and the risk and progression of malignancies. METHODS We searched the electronic databases for the studies regarding the relationship between tissue klotho protein expression and risk/progression of malignancies through January 2018. We calculated the pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) to evaluate the impact of tissue klotho protein expression on malignancies. A fixed-effect model, or in the presence of heterogeneity, random- effect model was applied to calculate the combined ORs. RESULTS Eighteen studies were recruited in our pooled-analysis. Overall malignancies including liver cancer, pancreatic ductal adenocarcinoma (PDAC), ovarian cancer, esophageal squamous cell carcinoma (ESCC), neuroendocrine cancer, oral cancer and bladder cancer demonstrated significantly lower ORs than those in controls (p < 0.05). Malignancies with tissue klotho protein expression showed a pooled hazard ratio (95% CI 0.784-2.479). Malignancies with tissue klotho protein expression showed a similar OR (95% CI 0.732-1.335) of male/total to cases without tissue klotho protein expression. Malignancies with tissue klotho protein expression showed a markedly lower OR (95% CI 0.454-0.941) of metastasis compared with those without tissue klotho protein expression. Malignancies with tissue klotho protein expression showed a markedly higher OR (95% CI 1.041-1.800) of stage I-II/III-IVcompared with those without tissue klotho protein expression. Malignancies with tissue klotho protein expression showed a similar OR (95% CI 0.948-3.407) of differentiation to cases without tissue klotho protein expression. Sensitivity analysis did not change the overall results significantly. No marked publication bias was noted. CONCLUSIONS Tissue klotho protein expression was associated with a lower risk and progression of malignancies. Klotho may be a protective factor against malignancies risk/progression.
Collapse
Affiliation(s)
- Song Mao
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200023, PR China
| | - Xiaopeng Wang
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200023, PR China
| | - Liangxia Wu
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200023, PR China
| | - Dou Zang
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200023, PR China
| | - Wenjing Shi
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200023, PR China.
| |
Collapse
|
21
|
Mencke R, Olauson H, Hillebrands JL. Effects of Klotho on fibrosis and cancer: A renal focus on mechanisms and therapeutic strategies. Adv Drug Deliv Rev 2017; 121:85-100. [PMID: 28709936 DOI: 10.1016/j.addr.2017.07.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/28/2017] [Accepted: 07/07/2017] [Indexed: 12/21/2022]
Abstract
Klotho is a membrane-bound protein predominantly expressed in the kidney, where it acts as a permissive co-receptor for Fibroblast Growth Factor 23. In its shed form, Klotho exerts anti-fibrotic effects in several tissues. Klotho-deficient mice spontaneously develop fibrosis and Klotho deficiency exacerbates the disease progression in fibrotic animal models. Furthermore, Klotho overexpression or supplementation protects against fibrosis in various models of renal and cardiac fibrotic disease. These effects are mediated at least partially by the direct inhibitory effects of soluble Klotho on TGFβ1 signaling, Wnt signaling, and FGF2 signaling. Soluble Klotho, as present in the circulation, appears to be the primary mediator of anti-fibrotic effects. Similarly, through inhibition of the TGFβ1, Wnt, FGF2, and IGF1 signaling pathways, Klotho also inhibits tumorigenesis. The Klotho promoter gene is generally hypermethylated in cancer, and overexpression or supplementation of Klotho has been found to inhibit tumor growth in various animal models. This review focuses on the protective effects of soluble Klotho in inhibiting renal fibrosis and fibrosis in distant organs secondary to renal Klotho deficiency. We also discuss the structure-function relationships of Klotho domains and biological effects in the context of potential targeted treatment strategies.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hannes Olauson
- Department of Clinical Science, Intervention and Technology (Division of Renal Medicine), Karolinska Institutet, Stockholm, Sweden
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
22
|
Mencke R, Harms G, Moser J, van Meurs M, Diepstra A, Leuvenink HG, Hillebrands JL. Human alternative Klotho mRNA is a nonsense-mediated mRNA decay target inefficiently spliced in renal disease. JCI Insight 2017; 2:94375. [PMID: 29046474 DOI: 10.1172/jci.insight.94375] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/14/2017] [Indexed: 12/11/2022] Open
Abstract
Klotho is a renal protein involved in phosphate homeostasis, which is downregulated in renal disease. It has long been considered an antiaging factor. Two Klotho gene transcripts are thought to encode membrane-bound and secreted Klotho. Indeed, soluble Klotho is detectable in bodily fluids, but the relative contributions of Klotho secretion and of membrane-bound Klotho shedding are unknown. Recent advances in RNA surveillance reveal that premature termination codons, as present in alternative Klotho mRNA (for secreted Klotho), prime mRNAs for degradation by nonsense-mediated mRNA decay (NMD). Disruption of NMD led to accumulation of alternative Klotho mRNA, indicative of normally continuous degradation. RNA IP for NMD core factor UPF1 resulted in enrichment for alternative Klotho mRNA, which was also not associated with polysomes, indicating no active protein translation. Alternative Klotho mRNA transcripts colocalized with some P bodies, where NMD transcripts are degraded. Moreover, we could not detect secreted Klotho in vitro. These results suggest that soluble Klotho is likely cleaved membrane-bound Klotho only. Furthermore, we found that, especially in acute kidney injury, splicing of the 2 mRNA transcripts is dysregulated, which was recapitulated by various noxious stimuli in vitro. This likely constitutes a novel mechanism resulting in the downregulation of membrane-bound Klotho.
Collapse
Affiliation(s)
- Rik Mencke
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| | - Geert Harms
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| | - Jill Moser
- Department of Intensive Care Medicine.,Department of Pathology and Medical Biology (Division of Medical Biology), and
| | - Matijs van Meurs
- Department of Intensive Care Medicine.,Department of Pathology and Medical Biology (Division of Medical Biology), and
| | - Arjan Diepstra
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Henri G Leuvenink
- Department of Surgery (Division of Experimental Surgery), University of Groningen, UMCG, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology (Division of Pathology), University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.,The NIGRAM consortium detailed in the Supplemental Acknowledgments
| |
Collapse
|
23
|
Olauson H, Mencke R, Hillebrands JL, Larsson TE. Tissue expression and source of circulating αKlotho. Bone 2017; 100:19-35. [PMID: 28323144 DOI: 10.1016/j.bone.2017.03.043] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/16/2022]
Abstract
αKlotho (Klotho), a type I transmembrane protein and a coreceptor for Fibroblast Growth Factor-23, was initially thought to be expressed only in a limited number of tissues, most importantly the kidney, parathyroid gland and choroid plexus. Emerging data may suggest a more ubiquitous Klotho expression pattern which has prompted reevaluation of the restricted Klotho paradigm. Herein we systematically review the evidence for Klotho expression in various tissues and cell types in humans and other mammals, and discuss potential reasons behind existing conflicting data. Based on current literature and tissue expression atlases, we propose a classification of tissues into high, intermediate and low/absent Klotho expression. The functional relevance of Klotho in organs with low expression levels remain uncertain and there is currently limited data on a role for membrane-bound Klotho outside the kidney. Finally, we review the evidence for the tissue source of soluble Klotho, and conclude that the kidney is likely to be the principal source of circulating Klotho in physiology.
Collapse
Affiliation(s)
- Hannes Olauson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Rik Mencke
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tobias E Larsson
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Yan Y, Wang Y, Xiong Y, Lin X, Zhou P, Chen Z. Reduced Klotho expression contributes to poor survival rates in human patients with ovarian cancer, and overexpression of Klotho inhibits the progression of ovarian cancer partly via the inhibition of systemic inflammation in nude mice. Mol Med Rep 2017; 15:1777-1785. [PMID: 28259911 DOI: 10.3892/mmr.2017.6172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 07/25/2016] [Indexed: 11/06/2022] Open
Abstract
Klotho is a recently discovered anti‑aging gene, which has been reported as a tumor suppressor in numerous human malignancies; however, the role of Klotho in human ovarian cancer remains to be elucidated. The aim of the present study was to detect the expression of Klotho and evaluate its association with the progression of human ovarian cancer. A clinical follow‑up study of 120 patients with ovarian cancer and 78 normal controls was conducted. The expression levels of Klotho were determined by western blotting and immunohistochemistry. The results demonstrated that high Klotho expression levels were detected in all normal controls, whereas the positive rate of Klotho was 61.6% in the ovarian cancer group, which was significantly decreased compared with in the control group (P<0.01). Furthermore, reduced Klotho expression was significantly correlated with decreased survival rates in patients with ovarian cancer (P=0.025). Subsequently, Klotho levels were detected in seven human ovarian cancer cell lines by western blotting. The results demonstrated that the highest levels of Klotho were detected in CaOV3 cells, medium levels of Klotho were detected in CaOV4 and SKOV‑3 cells, and almost no Klotho was detected in the other four cell lines: OVCA 432, OVCAR‑5, OVCAR‑8 and A2780 cells. The association between Klotho levels and cell proliferation was determined by MTT assay, and the results indicated that higher levels of Klotho inhibited the proliferation of A2780 and OVCAR‑5 cells, whereas reduced Klotho expression promoted cell growth of CaOV3 and SKOV‑3 cells. In addition, the plasma levels of inflammatory cytokines in tumor‑bearing mice and normal control mice were detected by enzyme‑linked immunosorbent assay, and plasma interleukin (IL)‑6 and IL‑1β levels were elevated in all tumor‑bearing mice. Notably, the mRNA expression levels of IL‑6 were significantly higher in the liver, ovaries and kidneys of Klotho‑/‑ mice compared with in wild type mice (P<0.01), thus indicating that aberrant Klotho expression may contribute to systemic inflammation in Klotho‑/‑ mice. Finally, the in vivo antitumor role of aberrant Klotho expression was determined in a nude mice model. A2780 cells were transfected with pCMV6‑Klotho and the stably transfected cells were screened; the mice were injected with the stably transfected cells. The results indicated that tumor volume and tumor weight were significantly decreased in the pCMV6‑Klotho group compared with in the pCMV6 vector group (P<0.01). These findings suggest that overexpression of Klotho may suppress tumor growth in animal models. In conclusion, Klotho was demonstrated to act as a potent tumor suppressor in human ovarian cancer cells. Reduced Klotho expression was detected in the specimens of patients with ovarian cancer, and overexpression of Klotho significantly inhibited cell proliferation of human ovarian cancer cells. Therefore, Klotho may be considered a useful key target for the molecular therapy of human ovarian cancer.
Collapse
Affiliation(s)
- Youliang Yan
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan of Southern Medical University, Guangzhou, Guangdong 528403, P.R. China
| | - Yifeng Wang
- Department of Obstetrics and Gynecology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Yi Xiong
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan of Southern Medical University, Guangzhou, Guangdong 528403, P.R. China
| | - Xiufeng Lin
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan of Southern Medical University, Guangzhou, Guangdong 528403, P.R. China
| | - Ping Zhou
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan of Southern Medical University, Guangzhou, Guangdong 528403, P.R. China
| | - Zhiying Chen
- Department of Obstetrics and Gynecology, Boai Hospital of Zhongshan of Southern Medical University, Guangzhou, Guangdong 528403, P.R. China
| |
Collapse
|
25
|
Dalton GD, Xie J, An SW, Huang CL. New Insights into the Mechanism of Action of Soluble Klotho. Front Endocrinol (Lausanne) 2017; 8:323. [PMID: 29250031 PMCID: PMC5715364 DOI: 10.3389/fendo.2017.00323] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022] Open
Abstract
The klotho gene encodes a type I single-pass transmembrane protein that contains a large extracellular domain, a membrane spanning segment, and a short intracellular domain. Klotho protein exists in several forms including the full-length membrane form (mKl) and a soluble circulating form [soluble klotho (sKl)]. mKl complexes with fibroblast growth factor receptors to form coreceptors for FGF23, which allows it to participate in FGF23-mediated signal transduction and regulation of phosphate and calcium homeostasis. sKl is present in the blood, urine, and cerebrospinal fluid where it performs a multitude of functions including regulation of ion channels/transporters and growth factor signaling. How sKl exerts these pleiotropic functions is poorly understood. One hurdle in understanding sKl's mechanism of action as a "hormone" has been the inability to identify a receptor that mediates its effects. In the body, the kidneys are a major source of sKl and sKl levels decline during renal disease. sKl deficiency in chronic kidney disease makes the heart susceptible to stress-induced injury. Here, we summarize the current knowledge of mKl's mechanism of action, the mechanistic basis of sKl's protective, FGF23-independent effects on the heart, and provide new insights into the mechanism of action of sKl focusing on recent findings that sKl binds sialogangliosides in membrane lipid rafts to regulate growth factor signaling.
Collapse
Affiliation(s)
- George D. Dalton
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, NC, United States
| | - Jian Xie
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sung-Wan An
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- *Correspondence: Chou-Long Huang,
| |
Collapse
|
26
|
Rubinek T, Wolf I. The Role of Alpha-Klotho as a Universal Tumor Suppressor. VITAMINS AND HORMONES 2016; 101:197-214. [PMID: 27125743 DOI: 10.1016/bs.vh.2016.03.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The klotho gene is implicated in many physiological activities, among them aging, glucose metabolism, and phosphate and calcium metabolism. Many cellular activities of klotho were implicated in promoting these activities. Two of them, inhibition of the insulin-like growth factor-1 pathway and of the Wnt signaling pathway, are also major pathways associated with cancer development and progression. These discoveries prompted a surge of research aiming to elucidate the role of klotho in cancer. Studies show that klotho is universally silenced in a wide array of malignancies, including breast, pancreatic, ovarian, lung, colorectal, and melanoma, and that klotho's expression can serve as an invaluable prognostic marker. Epigenetic mechanisms, ie, promoter hypermethylation and histone deacetylation, are mainly associated with klotho's silencing; however, different micro-RNAs were also demonstrated to be involved in the process. The activity of klotho on cancer cells growth was also widely investigated, and accumulating data suggest that klotho forced expression or treatment with the soluble protein can inhibit cancer development and progression. Moreover, studies now aim to reveal the specific region in klotho protein that underlies this anticancer activity in order to develop efficient and safe klotho-based medications.
Collapse
Affiliation(s)
- T Rubinek
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
| | - I Wolf
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
27
|
Sun H, Gao Y, Lu K, Zhao G, Li X, Li Z, Chang H. Overexpression of Klotho suppresses liver cancer progression and induces cell apoptosis by negatively regulating wnt/β-catenin signaling pathway. World J Surg Oncol 2015; 13:307. [PMID: 26499380 PMCID: PMC4619481 DOI: 10.1186/s12957-015-0717-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/05/2015] [Indexed: 01/30/2023] Open
Abstract
Background Klotho is a discovered aging suppressor gene, and its overexpression in mice extends the life span of the animal. Recently, Klotho is also identified as a tumor suppressor gene in variety of tumors; however, the potential role and the antitumor mechanism remain unclarified in liver cancers. Methods RT-PCR and western blotting analysis were used to detect the expression of Klotho, β-catenin, C-myc, and Cyclin D1. MTT assay was used to detect the survival rates of HepG2 and SMMC-7721 cells. Colony formation assay was used to test the proliferation ability in Klotho transfected cells. FACS was used to detect the cell apoptosis rate in different groups. Results The results showed that lower expression of Klotho were found in liver cancer cell lines than the immortalized liver cell L02. Also, MTT assay results found that overexpression or recombinant Klotho administration suppressed the proliferation of liver cancer cells HepG2 and SMMC-7721. Moreover, the colony formation assay results showed that the number of colonies was significantly lower in the cells with transfection with pCMV-Klotho than the controls. Thus, functional analysis demonstrated that Klotho expression inhibited the proliferation of liver cancer cells and Klotho worked as an important antitumor gene in tumor progression. Next, the mechanism was partly clarified that Klotho expression induced cell apoptosis in HepG2 and SMMC-7721 cells, and this phenomenon was mainly involved in the Wnt/β-catenin signaling pathway. The western blotting analysis revealed that overexpression or recombinant administration of Klotho obviously decreased the expression levels of β-catenin, C-myc, and Cyclin D1 in HepG2 cells. Most importantly, the antitumor mechanism for Klotho due to that overexpression of Klotho not only decreased the endogenous β-catenin levels but also inhibited the nuclear translocation of β-catenin to delay the cell cycle progression. Conclusions Klotho was a tumor suppressor gene, and overexpression of Klotho suppressed the proliferation of liver cancer cells partly due to negative regulation of Wnt/β-catenin signaling pathway. So, Klotho might be used as a potential target, and the study will contribute to treatment for therapy of liver cancer patients.
Collapse
Affiliation(s)
- Huidong Sun
- Department of Hepatobiliary, Surgery Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China.,Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China
| | - Yanchao Gao
- Department of Hepatobiliary, Surgery Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Kemei Lu
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Guimei Zhao
- Department of Hepatobiliary, Surgery Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Xuehua Li
- Department of Hepatobiliary, Surgery Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Zhu Li
- Department of Hepatobiliary, Surgery Liaocheng People's Hospital, Liaocheng, 252000, Shandong Province, China
| | - Hong Chang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong Province, China.
| |
Collapse
|