1
|
Tonini L, Ahn C. Latest Advanced Techniques for Improving Intestinal Organoids Limitations. Stem Cell Rev Rep 2025:10.1007/s12015-025-10894-9. [PMID: 40388043 DOI: 10.1007/s12015-025-10894-9] [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] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Intestinal organoids are valuable tools across different disciplines, from a clinical aspect to the biomedical research, providing a unique perspective on the complexity of the gastrointestinal system. They are alternatives to common cell lines as they can offer insights into architectural functionality and reduce the use of animal models. A deeper understanding of their organoid characteristics is required to harness their full potential. Despite their beneficial uses and multiple advantages, organoids have limitations that remain unaddressed. This review aims to elucidate the principal limitations of intestinal organoids, investigate structural defects such as the deficiency in a vascularized and lymphatic system, and absence of the microbiome, restrictions in mimicking the physiological gut model, including the lack of an acid-neutralizing system or a shortage of digestive enzymes, and the difficulties in their long-term maintenance and polarity accessibility. Development of innovative techniques to address these limitations will lead to improve in vivo recapitulation and pioneering further advancements in this field.
Collapse
Affiliation(s)
- Lisa Tonini
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea.
- Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
2
|
Wong GP, Hartmann S, Nonn O, Cannon P, Nguyen TV, Kandel M, de Alwis N, Murphy CN, Pritchard N, Dechend R, Hannan NJ, Tong S, Simmons DG, Kaitu'u-Lino TJ. Stem Cell Markers LGR5, LGR4 and Their Immediate Signalling Partners are Dysregulated in Preeclampsia. Stem Cell Rev Rep 2025; 21:872-896. [PMID: 39688759 DOI: 10.1007/s12015-024-10831-2] [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] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
Leucine-rich repeat-containing G protein-coupled receptors 5/4 (LGR5/LGR4) are critical stem cell markers in epithelial tissues including intestine. They agonise wingless-related integration site (WNT) signalling. Until now, LGR5/LGR4 were uncharacterised in placenta, where analogous functions may exist. We characterised LGR5/LGR4, their ligands/targets in human placenta, with further assessments on dysregulation in preeclampsia/fetal growth restriction (FGR). LGR5 mRNA was unaltered in first trimester (n = 11), preterm (n = 9) and term (n = 11) placental lysate. LGR5 was enriched in human trophoblast stem cells (hTSCs) and downregulated with differentiation to extravillous trophoblasts (p < 0.0215) and syncytiotrophoblasts (p < 0.0350). In situ hybridisation localised LGR5 to unique, proliferative MKI67 + mononuclear trophoblasts underlying syncytium which concurred with proposed progenitor identities in single-cell transcriptomics. LGR5 expression was significantly reduced in placentas from early-onset preeclampsia (p < 0.0001, n = 81 versus n = 19 controls), late-onset preeclampsia (p = 0.0046, n = 20 versus n = 33 controls) and FGR (p = 0.0031, n = 34 versus n = 17 controls). LGR4 was elevated in first trimester versus preterm and term placentas (p = 0.0412), in placentas with early-onset preeclampsia (p = 0.0148) and in FGR (p = 0.0417). Transcriptomic analysis and in vitro hTSC differentiation to both trophoblast lineages suggested LGR4 increases with differentiation. Single-nucleus RNA sequencing of placental villous samples supported LGR5 and LGR4 localisation findings. Hypoxia/proinflammatory cytokine treatment modelling elements experienced by the placenta in placental insufficiency pathogenesis did not significantly alter LGR5/LGR4. Ligands R-spondins 1/3/4, and neutralising targets ring finger protein 43 (RNF43) and zinc and ring finger 3 (ZNRF3) were also reduced in placentas from preeclamptic pregnancies. This study is the first to describe LGR5/LGR4 and their signalling partner expression in human placenta. Their dysregulations in the preeclamptic placenta allude to disruptions to integral trophoblast stem cell function/differentiation that may occur during placental development related to WNT signalling.
Collapse
Affiliation(s)
- Georgia P Wong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| | - Sunhild Hartmann
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
| | - Olivia Nonn
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ping Cannon
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Manju Kandel
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ciara N Murphy
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ralf Dechend
- Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and Charitè Campus Buch, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Klinikum, Berlin Buch, Germany
| | - Natalie J Hannan
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Stephen Tong
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - David G Simmons
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- The Department of Obstetrics, Gynaecology and Newborn Health/Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
3
|
Haque PS, Goodman D, Kuusivuori-Robinson T, Coughlan C, Delgado-Deida Y, Onyiah JC, Zempleni J, Theiss AL. Obese Adipose Tissue Extracellular Vesicles Activate Mitochondrial Fatty Acid β-oxidation to Drive Colonic Stemness. Cell Mol Gastroenterol Hepatol 2025; 19:101504. [PMID: 40122519 DOI: 10.1016/j.jcmgh.2025.101504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND & AIMS Patients with obesity and mouse models of obesity exhibit abnormalities in intestinal epithelial cells, including enhanced stemness. Adipose tissue (AT) is the largest endocrine organ secreting cytokines, hormones, and extracellular vesicles (EVs). Here, we characterized EV protein cargo from obese and non-obese AT and demonstrate the role of obese adipose-derived EVs in enhancing colonic stemness. METHODS EVs were isolated from visceral AT from mice fed high-fat diet to induce obesity or control matched-diet. EV cargo was characterized by unbiased proteomics. Mouse colonoids were treated with EVs and analyzed for fatty acid β-oxidation (FAO), expression of stem marker genes, stem function, and β-catenin expression and acetylation. Mice deficient in adipocyte-specific Tsg101 expression were generated to alter adipocyte EV protein cargo, and colonic stemness was measured. RESULTS EVs secreted from obese visceral AT (Ob EVs) were significantly enriched with acyl-CoA dehydrogenase long chain (ACADL), an initiator enzyme of FAO. Compared with non-obese EVs, colonoids treated with Ob EVs exhibited increased exogenous ACADL protein expression, FAO, growth, persistence of stem/progenitor function, and increased β-catenin protein expression and acetylation that was abolished by FAO inhibition. Mice deficient in adipocyte-specific Tsg101 expression exhibited Ob EVs with altered protein expression profiles and were protected from obesity-induced enhanced colonic stemness. CONCLUSIONS The contents of Ob EVs are poised to fuel FAO and to promote obesity-induced stemness in the colon. Alteration of metabolism is a key mechanism of adipose-to-intestinal tissue communication elicited by EVs, thereby influencing basal colonic stem cell homeostasis during obesity.
Collapse
Affiliation(s)
- Parsa S Haque
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Desiree Goodman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Thor Kuusivuori-Robinson
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Christina Coughlan
- Division of Neurology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Yaritza Delgado-Deida
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Joseph C Onyiah
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado.
| |
Collapse
|
4
|
Pun R, Thapa A, Takafuji SR, Suzuki RM, Kay GF, Howard TD, Kim MH, North BJ. BubR1 Controls Heart Development by Promoting Expression of Cardiogenesis Regulators. J Am Heart Assoc 2025; 14:e038286. [PMID: 40055864 DOI: 10.1161/jaha.124.038286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Congenital heart defects are structural anomalies present at birth that can affect the function of the heart. Aneuploidy is a significant risk factor for congenital heart defects. Mosaic variegated aneuploidy syndrome, caused by mutations in Bub1b (encoding BubR1, a mitotic checkpoint protein), leads to congenital heart defects such as septal defects. However, the molecular rationale for how Bub1b mutations promote congenital heart defects associated with mosaic variegated aneuploidy syndrome remains unresolved. METHODS To study morphological, structural, and cellular consequences of BubR1 deletion in the heart, we crossed mice carrying conditional alleles of Bub1b with Nkx2.5-cre mice. Single-cell RNA sequencing was carried out to determine differentially expressed genes and biological processes in various cell types present in the developing heart. Trajectory analysis was carried out to determine the differentiation trajectory of BubR1 knockout embryonic hearts. Finally, CellChat analysis provided details on the major signaling interactions that were either absent or hyperactive in the BubR1 knockout heart. RESULTS Here, we show that cardiac-specific BubR1 deletion causes embryonic lethality due to developmental stalling after cardiac looping with defects in cardiac maturation including chamber wall thickness, septation, and trabeculation. Single-cell transcriptomic profiling further revealed that the differentiation trajectory of cardiomyocytes is severely impacted with suppression of critical cardiogenesis genes. Hyperactivation of Wnt signaling in BubR1 knockout hearts indicated a disturbed homeostasis in cellular pathways essential for proper tissue morphogenesis of the heart. CONCLUSIONS Taken together, these findings reveal that BubR1 is a crucial regulator of cardiac development in vivo, which ensures the proper timing of heart morphogenesis.
Collapse
Affiliation(s)
- Renju Pun
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Aradhana Thapa
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Sylar R Takafuji
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Rexton M Suzuki
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Gabrielle F Kay
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Toni D Howard
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Michael H Kim
- CHI Heart Institute and Department of Medicine Creighton University School of Medicine Omaha NE USA
| | - Brian J North
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| |
Collapse
|
5
|
Wang W, Lokman NA, Barry SC, Oehler MK, Ricciardelli C. LGR5: An emerging therapeutic target for cancer metastasis and chemotherapy resistance. Cancer Metastasis Rev 2025; 44:23. [PMID: 39821694 PMCID: PMC11742290 DOI: 10.1007/s10555-024-10239-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
Cancer stem cells play an important role in tumor progression and chemotherapy resistance. Leucine-rich G repeat-containing protein-coupled receptor 5 (LGR5) has been identified as a cancer stem cell marker in several cancer types. LGR5 is involved in cancer development and progression via several pathways including WNT/β-catenin signaling pathway. LGR5 plays a role in tumor progression by promoting cancer cell migration, invasion, metastasis, and angiogenesis in many cancers including colorectal, brain, gastric, and ovarian cancer. This review summarises the current knowledge on the expression and functional role of LGR5 in cancers, the molecular mechanisms regulated by LGR5, and the relationship between LGR5 and chemotherapy resistance. The review also includes highlights potential strategies to inhibit LGR5 expression and function. The majority of functional studies have shown that LGR5 plays an important role in promoting cancer progression, metastasis and chemotherapy resistance however, in some contexts LGR5 can also activate tumor-suppressive pathways and LGR5 negative cells can also promote cancer progression. The review highlights that targeting LGR5 is a promising anti-cancer treatment but the functional effect of LGR5 on tumor cells is complex may be dependent on cancer type, tumor microenvironment and cross-talk with other molecules in the LGR5 signaling pathway.
Collapse
Affiliation(s)
- Wanqi Wang
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Noor A Lokman
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
| | - Simon C Barry
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, 5005, Australia
| | - Martin K Oehler
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, 5000, Australia
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, The University of Adelaide, Adelaide, 5005, Australia.
| |
Collapse
|
6
|
Hermo L, Oliveira R, Dufresne J, Gregory M, Cyr DG. Basal and Immune Cells of the Epididymis: An Electron Microscopy View of Their Association. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:67-87. [PMID: 40301253 DOI: 10.1007/978-3-031-82990-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The epididymis is a highly coiled duct divided into the initial segment, caput, corpus, and cauda regions. It is a pseudostratified epithelium consisting of principal, narrow, apical, basal, and clear cells. Circulating halo cells, identified as nonepithelial cells, monocytes/macrophages (M/M) and T-lymphocytes, in addition to dendritic cells and a resident population of M/M cells, also inhabit the epididymal epithelium. Using electron microscopy (EM), we characterized the ultrastructural features of each of these different cell types. Basal cells with stem cell characteristics suggest a role in sustaining the epithelium following injury and inflammation, as well as maintaining the steady state of the epithelium. Interestingly, a close morphological affiliation was noted between circulating M/M cells with basal cells and an intraepithelial resident M/M population of cells, as well as between T-lymphocytes and dendritic cells. The association of all these cell types with one another suggests complex interactions enabling the coordination of their functions related to maturation, protection, survival of sperm, and renewal of the epithelium.
Collapse
Affiliation(s)
- Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Regiana Oliveira
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | - Julie Dufresne
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Mary Gregory
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Daniel G Cyr
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada.
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada.
| |
Collapse
|
7
|
Nakamura K, Baba R, Kokubu K, Harada M, Morimoto H. Alterations in Ileal Secretory Cells of The DSS-Induced Colitis Model Mice. Acta Histochem Cytochem 2024; 57:199-209. [PMID: 39776935 PMCID: PMC11703563 DOI: 10.1267/ahc.24-00049] [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: 09/07/2024] [Accepted: 10/18/2024] [Indexed: 01/11/2025] Open
Abstract
Inflammatory bowel disease is triggered by abnormalities in epithelial barrier function and immunological responses, although its pathogenesis is poorly understood. The dextran sodium sulphate (DSS)-induced colitis model has been used to examine inflammation in the colon. Damage to mucosa primality occurs in the large intestine and scarcely in the small intestine. To evaluate the effect on the ileum, we histologically analyzed the inflammatory and recovery phases in DSS model mice, and 40 kDa FITC-dextran was used to investigate barrier function. In the inflammatory phase, histological damage was insignificant. However, expanded crypts, hypertrophic goblet and Paneth cells, increased mucus production and secretion were observed. The cellular morphology was restored to that of the control in the recovery phase. According to in situ hybridization and lectin histochemistry, the expression of intestinal stem cell markers, secretory cell differentiation factors, and glycosylation of secretory granules in Paneth cells differed in the DSS model. DSS-treatment did not influence the barrier function in the ileum, and FITC-dextran did not diffuse via the paracellular pathway into the mucosa. However, cells incorporating FITC appeared even under normal conditions. The number of FITC-positive Paneth cells was lower in the DSS group than the control group. Our results showed morphological and functional alterations in ileal epithelial cells, especially secretory cells, in the DSS colitis model.
Collapse
Affiliation(s)
- Kenta Nakamura
- Third Department of Internal Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Ryoko Baba
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Keiji Kokubu
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Masaru Harada
- Third Department of Internal Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| | - Hiroyuki Morimoto
- Department of Anatomy, School of Medicine, University of Occupational and Environmental Health, 1–1, Iseigaoka, Yahatanishi, Kitakyushu, Fukuoka 807–8555, Japan
| |
Collapse
|
8
|
Kappler M, Thielemann L, Glaß M, Caggegi L, Güttler A, Pyko J, Blauschmidt S, Gutschner T, Taubert H, Otto S, Eckert AW, Tavassol F, Bache M, Vordermark D, Kaune T, Rot S. Functional and Biological Characterization of the LGR5Δ5 Splice Variant in HEK293T Cells. Int J Mol Sci 2024; 25:13417. [PMID: 39769183 PMCID: PMC11678308 DOI: 10.3390/ijms252413417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
The regulator of the canonical Wnt pathway, leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), is expressed in the stem cell compartment of several tissues and overexpressed in different human carcinomas. The isoform of the stem cell marker LGR5, named LGR5Δ5 and first described by our group, is associated with prognosis and metastasis in oral squamous cell carcinoma (OSCC) and soft tissue sarcoma (STS). In a proof-of-principle analysis, the function of LGR5Δ5 was investigated in HEK293T cells, a model cell line of the Wnt pathway, compared to full-length LGR5 (FL) expression. The CRISPR/CAS knockout of LGR5 and LGR4 (thereby avoiding the side effects of LGR4) resulted in a loss of Wnt activity that cannot be restored by LGR5Δ5 but by LGR5FL rescue. The ability to migrate was not affected by LGR5Δ5, but was reduced by LGR5FL overexpression. The CRISPR/CAS of LGR4 and 5 induced radiosensitization, which was enhanced by the overexpression of LGR5FL or LGR5Δ5. RNA sequencing analysis revealed a significant increase in the ligand R-spondin 1 (RSPO1) level by LGR5Δ5. Furthermore, LGR5Δ5 appears to be involved in the regulation of genes related to the cytoskeleton, extracellular matrix stiffness, and angiogenesis, while LGR5FL is associated with the regulation of collagens and histone proteins.
Collapse
Affiliation(s)
- Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Laura Thielemann
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Markus Glaß
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany;
| | - Laura Caggegi
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Antje Güttler
- Department of Radiotherapy, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (A.G.); (M.B.); (D.V.)
| | - Jonas Pyko
- Institute of Molecular Medicine, Section for RNA Biology and Pathogenesis, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (J.P.); (T.G.)
| | - Sarah Blauschmidt
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Tony Gutschner
- Institute of Molecular Medicine, Section for RNA Biology and Pathogenesis, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (J.P.); (T.G.)
| | - Helge Taubert
- Department of Urology, University Hospital Erlangen, Friedrich-Alexander-University of Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Sven Otto
- Department of Oral and Maxillofacial Surgery and Facial Plastic Surgery, Ludwig Maximilians University, 80337 Munich, Germany;
| | - Alexander W. Eckert
- Department of Cranio Maxillofacial Surgery, Paracelsus Medical University, 90471 Nuremberg, Germany;
| | - Frank Tavassol
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Matthias Bache
- Department of Radiotherapy, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (A.G.); (M.B.); (D.V.)
| | - Dirk Vordermark
- Department of Radiotherapy, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (A.G.); (M.B.); (D.V.)
| | - Tom Kaune
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| | - Swetlana Rot
- Department of Oral and Maxillofacial Plastic Surgery, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany (S.B.); (F.T.)
| |
Collapse
|
9
|
Schöpe PC, Torke S, Kobelt D, Kortüm B, Treese C, Dumbani M, Güllü N, Walther W, Stein U. MACC1 revisited - an in-depth review of a master of metastasis. Biomark Res 2024; 12:146. [PMID: 39580452 PMCID: PMC11585957 DOI: 10.1186/s40364-024-00689-4] [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: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cancer metastasis remains the most lethal characteristic of tumors mediating the majority of cancer-related deaths. Identifying key molecules responsible for metastasis, understanding their biological functions and therapeutically targeting these molecules is therefore of tremendous value. Metastasis Associated in Colon Cancer 1 (MACC1), a gene first described in 2009, is such a key driver of metastatic processes, initiating cellular proliferation, migration, invasion, and metastasis in vitro and in vivo. Since its discovery, the value of MACC1 as a prognostic biomarker has been confirmed in over 20 cancer entities. Additionally, several therapeutic strategies targeting MACC1 and its pro-metastatic functions have been developed. In this review, we will provide a comprehensive overview on MACC1, from its clinical relevance, towards its structure and role in signaling cascades as well as molecular networks. We will highlight specific biological consequences of MACC1 expression, such as an increase in stem cell properties, its immune-modulatory effects and induced therapy resistance. Lastly, we will explore various strategies interfering with MACC1 expression and/or its functions. Conclusively, this review underlines the importance of understanding the role of individual molecules in mediating metastasis.
Collapse
Affiliation(s)
- Paul Curtis Schöpe
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Sebastian Torke
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Benedikt Kortüm
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Christoph Treese
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Malti Dumbani
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Nazli Güllü
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
- German Cancer Consortium (DKTK), Berlin and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
10
|
Uddin J, Sharma A, Wu D, Tomar S, Ganesan V, Reichel PE, Thota LNR, Cabrera-Silva RI, Marella S, Idelman G, Tay HL, Raya-Sandino A, Reynolds MB, Elesela S, Haberman Y, Denson LA, Parkos CA, O’Riordan MX, Lukacs NW, O’Dwyer DN, Divanovic S, Nusrat A, Weaver TE, Hogan SP. STARD7 maintains intestinal epithelial mitochondria architecture, barrier integrity, and protection from colitis. JCI Insight 2024; 9:e172978. [PMID: 39576011 PMCID: PMC11601949 DOI: 10.1172/jci.insight.172978] [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: 06/12/2023] [Accepted: 09/25/2024] [Indexed: 11/29/2024] Open
Abstract
Susceptibility to inflammatory bowel diseases (IBDs), Crohn's disease (CD), and ulcerative colitis (UC) is linked with loss of intestinal epithelial barrier integrity and mitochondria dysfunction. Steroidogenic acute regulatory (StAR) protein-related lipid transfer (START) domain-containing protein 7 (STARD7) is a phosphatidylcholine-specific (PC-specific) lipid transfer protein that transports PC from the ER to the mitochondria, facilitating mitochondria membrane stabilization and respiration function. The aim of this study was to define the contribution of STARD7 in the regulation of the intestinal epithelial mitochondrial function and susceptibility to colitis. In silico analyses identified significantly reduced expression of STARD7 in patients with UC, which was associated with downregulation of metabolic function and a more severe disease phenotype. STARD7 was expressed in intestinal epithelial cells, and STARD7 knockdown resulted in deformed mitochondria and diminished aerobic respiration. Loss of mitochondria function was associated with reduced expression of tight junction proteins and loss of intestinal epithelial barrier integrity that could be recovered by AMPK activation. Stard7+/- mice were more susceptible to the development of DSS-induced and Il10-/- spontaneous models of colitis. STARD7 is critical for intestinal epithelial mitochondrial function and barrier integrity, and loss of STARD7 function increases susceptibility to IBD.
Collapse
Affiliation(s)
- Jazib Uddin
- Division of Experimental Pathology, Department of Pathology, and
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ankit Sharma
- Division of Experimental Pathology, Department of Pathology, and
| | - David Wu
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sunil Tomar
- Division of Experimental Pathology, Department of Pathology, and
| | - Varsha Ganesan
- Division of Experimental Pathology, Department of Pathology, and
| | - Paula E. Reichel
- Division of Experimental Pathology, Department of Pathology, and
| | | | | | - Sahiti Marella
- Division of Experimental Pathology, Department of Pathology, and
| | - Gila Idelman
- Division of Experimental Pathology, Department of Pathology, and
| | - Hock L. Tay
- Division of Experimental Pathology, Department of Pathology, and
| | | | - Mack B. Reynolds
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Srikanth Elesela
- Division of Experimental Pathology, Department of Pathology, and
| | - Yael Haberman
- Sheba Medical Center, Tel-Hashomer, and
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lee A. Denson
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | | | - Mary X.D. O’Riordan
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nicholas W. Lukacs
- Division of Experimental Pathology, Department of Pathology, and
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David N. O’Dwyer
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Senad Divanovic
- Division of Immunobiology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance and
| | - Asma Nusrat
- Division of Experimental Pathology, Department of Pathology, and
| | - Timothy E. Weaver
- Divisions of Neonatology, Perinatal Biology, and Pulmonary Biology, Perinatal Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Simon P. Hogan
- Division of Experimental Pathology, Department of Pathology, and
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Trubin S, Patel DB, Tian A. Regulation of the Intestinal Stem Cell Pool and Proliferation in Drosophila. Cells 2024; 13:1856. [PMID: 39594605 PMCID: PMC11592481 DOI: 10.3390/cells13221856] [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: 10/23/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Understanding the regulation of somatic stem cells, both during homeostasis and in response to environmental challenges like injury, infection, chemical exposure, and nutritional changes, is critical because their dysregulation can result in tissue degeneration or tumorigenesis. The use of models such as the Drosophila and mammalian adult intestines offers valuable insights into tissue homeostasis and regeneration, advancing our knowledge of stem cell biology and cancer development. This review highlights significant findings from recent studies, unveiling the molecular mechanisms that govern self-renewal, proliferation, differentiation, and regeneration of intestinal stem cells (ISCs). These insights not only enhance our understanding of normal tissue maintenance but also provide critical perspectives on how ISC dysfunction can lead to pathological conditions such as colorectal cancer (CRC).
Collapse
Affiliation(s)
- Simona Trubin
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Dhruv B. Patel
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Aiguo Tian
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
- Tulane Aging Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
12
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
13
|
Tu J, Toh Y, Aldana AM, Wen JJ, Wu L, Jacob J, Li L, Pan S, Carmon KS, Liu QJ. Antitumor Activity of a Pyrrolobenzodiazepine Antibody-Drug Conjugate Targeting LGR5 in Preclinical Models of Neuroblastoma. Pharmaceutics 2024; 16:943. [PMID: 39065640 PMCID: PMC11279891 DOI: 10.3390/pharmaceutics16070943] [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: 06/20/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Neuroblastoma (NB) is a cancer of the peripheral nervous system found in children under 15 years of age. It is the most frequently diagnosed cancer during infancy, accounting for ~12% of all cancer-related deaths in children. Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) is a membrane receptor that is associated with the primary tumor formation and metastasis of cancers in the gastrointestinal system. Remarkably, high levels of LGR5 are found in NB tumor cells, and high LGR5 expression is strongly correlated with poor survival. Antibody-drug conjugates (ADCs) are monoclonal antibodies that are covalently linked to cell-killing cytotoxins to deliver the payloads into cancer cells. We generated an ADC with an anti-LGR5 antibody and pyrrolobenzodiazepine (PBD) dimer-based payload SG3199 using a chemoenzymatic conjugation method. The resulting anti-LGR5 ADC was able to inhibit the growth of NB cells expressing LGR5 with high potency and specificity. Importantly, the ADC was able to completely inhibit the growth of NB xenograft tumors in vivo at a clinically relevant dose for the PBD class of ADCs. The findings support the potential of targeting LGR5 using the PBD class of payload for the treatment of high-risk NBs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Qingyun J. Liu
- The Brown Foundation Institute of Molecular Medicine, Center for Translational Cancer Research, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
14
|
Shan X, Rathore S, Kniffen D, Gao L, Nitin, Letef CL, Shi H, Ghosh S, Zandberg W, Xia L, Bergstrom KS. Ablation of Intestinal Epithelial Sialylation Predisposes to Acute and Chronic Intestinal Inflammation in Mice. Cell Mol Gastroenterol Hepatol 2024; 18:101378. [PMID: 38992465 PMCID: PMC11459652 DOI: 10.1016/j.jcmgh.2024.101378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND & AIMS Addition of sialic acids (sialylation) to glycoconjugates is a common capping step of glycosylation. Our study aims to determine the roles of the overall sialylation in intestinal mucosal homeostasis. METHODS Mice with constitutive deletion of intestinal epithelial sialylation (IEC Slc35a1-/- mice) and mice with inducible deletion of sialylation in intestinal epithelium (TM-IEC Slc35a1-/- mice) were generated, which were used to determine the roles of overall sialylation in intestinal mucosal homeostasis by ex vivo and mutiomics studies. RESULTS IEC Slc35a1-/- mice developed mild spontaneous microbiota-dependent colitis. Additionally, 30% of IEC Slc35a1-/- mice had spontaneous tumors in the rectum greater than the age of 12 months. TM-IEC Slc35a1-/- mice were highly susceptible to acute inflammation induced by 1% dextran sulfate sodium versus control animals. Loss of total sialylation was associated with reduced mucus thickness on fecal sections and within colon tissues. TM-IEC Slc35a1-/- mice showed altered microbiota with an increase in Clostridium disporicum, which is associated a global reduction in the abundance of at least 10 unique taxa; however, metabolomic analysis did not show any significant differences in short-chain fatty acid levels. Treatment with 5-fluorouracil led to more severe small intestine mucositis in the IEC Slc35a1-/- mice versus wild-type littermates, which was associated with reduced Lgr5+ cell representation in small intestinal crypts in IEC Slc35a1-/-;Lgr5-GFP mice. CONCLUSIONS Loss of overall sialylation impairs mucus stability and the stem cell niche leading to microbiota-dependent spontaneous colitis and tumorigenesis.
Collapse
Affiliation(s)
- Xindi Shan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Shipra Rathore
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Darrek Kniffen
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Liang Gao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Nitin
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Clara L Letef
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Huiping Shi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Sanjoy Ghosh
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Wesley Zandberg
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Kirk S Bergstrom
- Department of Biology, University of British Columbia, Okanagan Campus, Kelowna, British Columbia, Canada.
| |
Collapse
|
15
|
Hill ABT, Murphy YM, Polkoff KM, Edwards L, Walker DM, Moatti A, Greenbaum A, Piedrahita JA. A gene edited pig model for studying LGR5 + stem cells: implications for future applications in tissue regeneration and biomedical research. Front Genome Ed 2024; 6:1401163. [PMID: 38903529 PMCID: PMC11187295 DOI: 10.3389/fgeed.2024.1401163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Recent advancements in genome editing techniques, notably CRISPR-Cas9 and TALENs, have marked a transformative era in biomedical research, significantly enhancing our understanding of disease mechanisms and helping develop novel therapies. These technologies have been instrumental in creating precise animal models for use in stem cell research and regenerative medicine. For instance, we have developed a transgenic pig model to enable the investigation of LGR5-expressing cells. The model was designed to induce the expression of H2B-GFP under the regulatory control of the LGR5 promoter via CRISPR/Cas9-mediated gene knock-in. Notably, advancements in stem cell research have identified distinct subpopulations of LGR5-expressing cells within adult human, mouse, and pig tissues. LGR5, a leucine-rich repeat-containing G protein-coupled receptor, enhances WNT signaling and these LGR5+ subpopulations demonstrate varied roles and anatomical distributions, underscoring the necessity for suitable translational models. This transgenic pig model facilitates the tracking of LGR5-expressing cells and has provided valuable insights into the roles of these cells across different tissues and species. For instance, in pulmonary tissue, Lgr5+ cells in mice are predominantly located in alveolar compartments, driving alveolar differentiation of epithelial progenitors via Wnt pathway activation. In contrast, in pigs and humans, these cells are situated in a unique sub-basal position adjacent to the airway epithelium. In fetal stages a pattern of LGR5 expression during lung bud tip formation is evident in humans and pigs but is lacking in mice. Species differences with respect to LGR5 expression have also been observed in the skin, intestines, and cochlea further reinforcing the need for careful selection of appropriate translational animal models. This paper discusses the potential utility of the LGR5+ pig model in exploring the role of LGR5+ cells in tissue development and regeneration with the goal of translating these findings into human and animal clinical applications.
Collapse
Affiliation(s)
- Amanda B. T. Hill
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Yanet M. Murphy
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Kathryn M. Polkoff
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Laura Edwards
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Derek M. Walker
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| | - Jorge A. Piedrahita
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
16
|
Miao D, Ren J, Jia Y, Jia Y, Li Y, Huang H, Gao R. PAX1 represses canonical Wnt signaling pathway and plays dual roles during endoderm differentiation. Cell Commun Signal 2024; 22:242. [PMID: 38664733 PMCID: PMC11046865 DOI: 10.1186/s12964-024-01629-3] [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: 01/02/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Paired box 1 (PAX1) is a transcription factor and essential for the development of pharyngeal pouches-derived tissues, including thymus. PAX1 mutations are identified in Severe Combined Immunodeficiency (SCID) patients with Otofaciocervical Syndrome Type 2 (OTFCS2). However, despite the critical roles of PAX1 in embryonic development and diseases, detailed insights into its molecular mode of action are critically missing. METHODS The repressing roles of PAX1 and SCID associated mutants on Wnt signaling pathway were investigated by luciferase reporter assays, qRT-PCR and in situ hybridization in HEK293FT, HCT116 cells and zebrafish embryos, respectively. Co-immunoprecipitation (co-IP) and western blotting assays were carried out to identify the molecular mechanisms underlying PAX1's role on Wnt signaling pathway. hESC based endoderm differentiation, flow cytometry, high-throughput sequencing data analysis, and qRT-PCR assays were utilized to determine the roles of PAX1 during endoderm differentiation. RESULTS Here, we show that PAX1 represses canonical Wnt signaling pathway in vertebrate cells. Mechanically, PAX1 competes with SUMO E3 ligase PIASy to bind to TCF7L2, thus perturbing TCF7L2 SUMOylation level, further reducing its transcriptional activity and protein stability. Moreover, we reveal that PAX1 plays dual roles in hESC-derived definitive and foregut/pharyngeal endoderm cells, which give rise to the thymus epithelium, by inhibiting Wnt signaling. Importantly, our data show PAX1 mutations found in SCID patients significantly compromise the suppressing ability of PAX1 on Wnt signaling. CONCLUSIONS Our study presents a novel molecular mode of action of PAX1 in regulation of canonical Wnt signaling and endoderm differentiation, thus providing insights for the molecular basis of PAX1 associated SCID, offering better understanding of the behavior of PAX1 in embryogenesis.
Collapse
Affiliation(s)
- Danxiu Miao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, 150000, China
| | - Jie Ren
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
| | - Yanhan Jia
- Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China
| | - Yihui Jia
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China
| | - Yanshu Li
- Department of Toxicology, College of Public Health, Harbin Medical University, Harbin, 150000, China
- College of Public Health, Shantou University, Shantou, 515063, China
| | - Huizhe Huang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Rui Gao
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of medicine, Xiamen University, Xiamen, 361000, China.
| |
Collapse
|
17
|
Jiang KL, Jia YB, Liu XJ, Jia QL, Guo LK, Wang XX, Yang KM, Wu CH, Liang BB, Ling JH. Bibliometrics analysis based on the Web of Science: Current trends and perspective of gastric organoid during 2010-2023. World J Gastroenterol 2024; 30:969-983. [PMID: 38516239 PMCID: PMC10950634 DOI: 10.3748/wjg.v30.i8.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/02/2024] [Accepted: 02/01/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Three-dimensional organoid culture systems have been established as a robust tool for elucidating mechanisms and performing drug efficacy testing. The use of gastric organoid models holds significant promise for advancing personalized medicine research. However, a comprehensive bibliometric review of this bur-geoning field has not yet been published. AIM To analyze and understand the development, impact, and direction of gastric organoid research using bibliometric methods using data from the Web of Science Core Collection (WoSCC) database. METHODS This analysis encompassed literature pertaining to gastric organoids published between 2010 and 2023, as indexed in the WoSCC. CiteSpace and VOSviewer were used to depict network maps illustrating collaborations among authors, institutions and keywords related to gastric organoid. Citation, co-citation, and burst analysis methodologies were applied to assess the impact and progress of research. RESULTS A total of 656 relevant studies were evaluated. The majority of research was published in gastroenterology-focused journals. Globally, Yana Zavros, Hans Clevers, James M Wells, Sina Bartfeld, and Chen Zheng were the 5 most productive authors, while Hans Clevers, Huch Meritxell, Johan H van Es, Marc Van de Wetering, and Sato Toshiro were the foremost influential scientists in this area. Institutions from the University Medical Center Utrecht, Netherlands Institute for Developmental Biology (Utrecht), and University of Cincinnati (Cincinnati, OH, United States) made the most significant contributions. Currently, gastric organoids are used mainly in studies investigating gastric cancer (GC), Helicobacter pylori-infective gastritis, with a focus on the mechanisms of GC, and drug screening tests. CONCLUSION Key focus areas of research using gastric organoids include unraveling disease mechanisms and enhancing drug screening techniques. Major contributions from renowned academic institutions highlight this field's dynamic growth.
Collapse
Affiliation(s)
- Kai-Lin Jiang
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
- Laboratory of Cancer Biology, University of Oxford, Oxford OX37DQ, United Kingdom
| | - Yue-Bo Jia
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xue-Jiao Liu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Qing-Ling Jia
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Li-Kun Guo
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Xiang-Xiang Wang
- School of Traditional Chinese Medicine, Shanghai University of Chinese Medicine, Shanghai 200021, China
| | - Ke-Ming Yang
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Chen-Heng Wu
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Bei-Bei Liang
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jiang-Hong Ling
- Department of Gastroenterology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| |
Collapse
|
18
|
Erdem M, Lee KH, Hardt M, Regan JL, Kobelt D, Walther W, Mokrizkij M, Regenbrecht C, Stein U. MACC1 Regulates LGR5 to Promote Cancer Stem Cell Properties in Colorectal Cancer. Cancers (Basel) 2024; 16:604. [PMID: 38339354 PMCID: PMC10854991 DOI: 10.3390/cancers16030604] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. The high mortality is directly associated with metastatic disease, which is thought to be initiated by colon cancer stem cells, according to the cancer stem cell (CSC) model. Consequently, early identification of those patients who are at high risk for metastasis is crucial for improved treatment and patient outcomes. Metastasis-associated in colon cancer 1 (MACC1) is a novel prognostic biomarker for tumor progression and metastasis formation independent of tumor stage. We previously showed an involvement of MACC1 in cancer stemness in the mouse intestine of our MACC1 transgenic mouse models. However, the expression of MACC1 in human CSCs and possible implications remain elusive. Here, we explored the molecular mechanisms by which MACC1 regulates stemness and the CSC-associated invasive phenotype based on patient-derived tumor organoids (PDOs), patient-derived xenografts (PDXs) and human CRC cell lines. We showed that CD44-enriched CSCs from PDO models express significantly higher levels of MACC1 and LGR5 and display higher tumorigenicity in immunocompromised mice. Similarly, RNA sequencing performed on PDO and PDX models demonstrated significantly increased MACC1 expression in ALDH1(+) CSCs, highlighting its involvement in cancer stemness. We further showed the correlation of MACC1 with the CSC markers CD44, NANOG and LGR5 in PDO models as well as established cell lines. Additionally, MACC1 increased stem cell gene expression, clonogenicity and sphere formation. Strikingly, we showed that MACC1 binds as a transcription factor to the LGR5 gene promoter, uncovering the long-known CSC marker LGR5 as a novel essential signaling mediator employed by MACC1 to induce CSC-like properties in human CRC patients. Our in vitro findings were further substantiated by a significant positive correlation of MACC1 with LGR5 in CRC cell lines as well as CRC patient tumors. Taken together, this study indicates that the metastasis inducer MACC1 acts as a cancer stem cell-associated marker. Interventional approaches targeting MACC1 would potentially improve further targeted therapies for colorectal cancer patients to eradicate CSCs and prevent cancer recurrence and distant metastasis formation.
Collapse
Affiliation(s)
- Müge Erdem
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
| | - Kyung Hwan Lee
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
| | - Markus Hardt
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
| | - Joseph L. Regan
- Bayer AG, Research and Development, Pharmaceuticals, 13342 Berlin, Germany
- JLR Life Sciences Ltd., A96 A8D5 Dublin, Ireland
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
- German Cancer Consortium, 69120 Heidelberg, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
| | - Margarita Mokrizkij
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
| | | | - Ulrike Stein
- Experimental and Clinical Research Center, Charité—Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Translational Oncology of Solid Tumors Research Group, 13125 Berlin, Germany (D.K.)
- German Cancer Consortium, 69120 Heidelberg, Germany
| |
Collapse
|
19
|
Zhang Y, Chen W, Dong X, Shang W, Shao S, Zhang L. Long-term maintenance of human endometrial epithelial organoids and their stem cell properties. Reprod Toxicol 2024; 123:108522. [PMID: 38096957 DOI: 10.1016/j.reprotox.2023.108522] [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: 10/09/2023] [Revised: 11/17/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
The endometrium undergoes dynamic changes throughout the menstrual cycle and pregnancy, which is unique to primates. Endometrium remodeling is essential for the implantation and nutritional support of the conceptus. Despite this, the role of uterine glands in driving endometrial tissue remodeling is still poorly understood. To address this, a 3-dimensional culture system was used to generate endometrial epithelial organoids from human endometrium biopsies. These organoids are genetically stable, long-term expandability. They reproduce some functions of uterine glands in vivo. The epithelial organoids exhibit characteristics of stem cells, with the proportion of stem cells increasing with culture time and passage number. Long-term maintenance of organoids strongly expressed stemness related genes accompanied by a decrease expression in mature epithelial gene, which suggests the organoids had switched from a mature stage to a progenitor stage. Thus we proposed the possible markers for epithelial progenitors. Meanwhile, long-term cultured organoids exhibit an increase in the proportion of luminal epithelial stem cells, accompanied by a decrease of glandular epithelial stem cells. Organoids also show hormone responsiveness, reflecting the various stages of the menstrual cycle and early pregnancy.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Histology and Embryology, Hebei Medical University, 050000 Shijiazhuang, China
| | - Wei Chen
- Department of Histology and Embryology, Hebei Medical University, 050000 Shijiazhuang, China
| | - Xiaomin Dong
- Medical School of Chinese People's Liberation Army (PLA), 100010 Beijing, China
| | - Wei Shang
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, 100010 Beijing, China
| | - Suxia Shao
- Department of Histology and Embryology, Hebei Medical University, 050000 Shijiazhuang, China
| | - Lei Zhang
- Department of Histology and Embryology, Hebei Medical University, 050000 Shijiazhuang, China.
| |
Collapse
|
20
|
Torborg SR, Grbovic-Huezo O, Singhal A, Holm M, Wu K, Han X, Ho YJ, Haglund C, Mitchell MJ, Lowe SW, Dow LE, Pitter KL, Sanchez-Rivera FJ, Levchenko A, Tammela T. Solid tumor growth depends on an intricate equilibrium of malignant cell states. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.30.573100. [PMID: 38234855 PMCID: PMC10793475 DOI: 10.1101/2023.12.30.573100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Control of cell identity and number is central to tissue function, yet principles governing organization of malignant cells in tumor tissues remain poorly understood. Using mathematical modeling and candidate-based analysis, we discover primary and metastatic pancreatic ductal adenocarcinoma (PDAC) organize in a stereotypic pattern whereby PDAC cells responding to WNT signals (WNT-R) neighbor WNT-secreting cancer cells (WNT-S). Leveraging lineage-tracing, we reveal the WNT-R state is transient and gives rise to the WNT-S state that is highly stable and committed to organizing malignant tissue. We further show that a subset of WNT-S cells expressing the Notch ligand DLL1 form a functional niche for WNT-R cells. Genetic inactivation of WNT secretion or Notch pathway components, or cytoablation of the WNT-S state disrupts PDAC tissue organization, suppressing tumor growth and metastasis. This work indicates PDAC growth depends on an intricately controlled equilibrium of functionally distinct cancer cell states, uncovering a fundamental principle governing solid tumor growth and revealing new opportunities for therapeutic intervention.
Collapse
|
21
|
Wang J, Koch DT, Hofmann FO, Härtwig D, Beirith I, Janssen KP, Bazhin AV, Niess H, Werner J, Renz BW, Ilmer M. WNT enhancing signals in pancreatic cancer are transmitted by LGR6. Aging (Albany NY) 2023; 15:10897-10914. [PMID: 37770230 PMCID: PMC10637827 DOI: 10.18632/aging.205101] [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: 04/03/2023] [Accepted: 07/17/2023] [Indexed: 10/03/2023]
Abstract
The G-protein-coupled receptor LGR6 associates with ligands of the R-Spondin (RSPO) family to potentiate preexisting signals of the canonical WNT pathway. However, its importance in pancreatic ductal adenocarcinoma (PDAC) remains unclear. Here, we show that LGR6 is differentially expressed in various PDAC cell lines of mesenchymal and epithelial phenotype, respectively, siding with the latter subsets. LGR6 expression is altered based upon the cells' WNT activation status. Furthermore, extrinsic enhancement of WNT pathway signaling increased LGR6 expression suggestive of a reinforcing self-regulatory loop in highly WNT susceptible cells. Downregulation of LGR6 on the other hand, seemed to tamper those effects. Last, downregulation of LGR6 reduced cancer stemness as determined by functional in vitro assays. These findings shed new insights into regulatory mechanisms for the canonical WNT pathway in pancreatic cancer cells. It may also have potential value for treatment stratification of PDAC.
Collapse
Affiliation(s)
- Jing Wang
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P.R. China
| | - Dominik T. Koch
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
| | - Felix O. Hofmann
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
| | - Daniel Härtwig
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
| | - Iris Beirith
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
| | - Klaus Peter Janssen
- Department of Surgery, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Bavaria, Germany
| | - Alexandr V. Bazhin
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Bavaria, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Munich, Bavaria, Germany
| | - Hanno Niess
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
| | - Jens Werner
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Bavaria, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Munich, Bavaria, Germany
| | - Bernhard W. Renz
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Bavaria, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Munich, Bavaria, Germany
| | - Matthias Ilmer
- Department of General, Visceral and Transplantation Surgery, Hospital of the University of Munich, Ludwig-Maximilians-University (LMU), Munich, Bavaria, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Bavaria, Germany
- Bavarian Cancer Research Center (BZKF), LMU Munich, Munich, Bavaria, Germany
| |
Collapse
|
22
|
Jamal Eddin TM, Nasr SM, Gupta I, Zayed H, Al Moustafa AE. Helicobacter pylori and epithelial mesenchymal transition in human gastric cancers: An update of the literature. Heliyon 2023; 9:e18945. [PMID: 37609398 PMCID: PMC10440535 DOI: 10.1016/j.heliyon.2023.e18945] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Gastric cancer, a multifactorial disease, is considered one of the most common malignancies worldwide. In addition to genetic and environmental risk factors, infectious agents, such as Epstein-Barr virus (EBV) and Helicobacter pylori (H.pylori) contribute to the onset and development of gastric cancer. H. pylori is a type I carcinogen that colonizes the gastric epithelium of approximately 50% of the world's population, thus increasing the risk of gastric cancer development. On the other hand, epithelial mesenchymal transition (EMT) is a fundamental process crucial to embryogenic growth, wound healing, organ fibrosis and cancer progression. Several studies associate gastric pathogen infection of the epithelium with EMT initiation, provoking cancer metastasis in the gastric mucosa through various molecular signaling pathways. Additionally, EMT is implicated in the progression and development of H. pylori-associated gastric cancer. In this review, we recapitulate recent findings elucidating the association between H. pylori infection in EMT promotion leading to gastric cancer progression and metastasis.
Collapse
Affiliation(s)
- Tala M. Jamal Eddin
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Shahd M.O. Nasr
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hatem Zayed
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical Research Center, Qatar University, PO Box 2713, Doha, Qatar
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
23
|
Toh Y, Wu L, Park S, Wang A, Tu J, Yu W, Zuo M, Carmon KS, Liu QJ. LGR4 and LGR5 form distinct homodimers that only LGR4 complexes with RNF43/ZNRF3 to provide high affinity binding of R-spondin ligands. Sci Rep 2023; 13:10796. [PMID: 37402772 DOI: 10.1038/s41598-023-37856-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/28/2023] [Indexed: 07/06/2023] Open
Abstract
LGR4 and LGR5 are two homologous receptors that potentiate Wnt/β-catenin signaling in response to R-spondin (RSPO) ligands. The RSPO and LGR4 complex binds to and inhibits activities of two related E3 ubiquitin ligases, RNF43 and ZNRF3, and thus protects Wnt receptors from the E3 ligase-mediated degradation. The RSPO and LGR5 complex, however, does not interact with the E3 ligases, and the structural basis of this difference remained unknown. Here we examined the affinities of monovalent and bivalent RSPO ligands in binding to LGR4, RNF43/ZNRF3, and LGR5 in whole cells and found unique features among the receptors and E3 ligases. Monovalent RSPO2 furin domain had much lower affinity in binding to LGR4 or RNF43/ZNRF3 than the bivalent form. In contrast, monovalent and bivalent forms had nearly identical affinity in binding to LGR5. Co-expression of ZNRF3 with LGR4 led to much higher binding affinity of the monovalent form whereas co-expression of ZNRF3 with LGR5 had no effect on the affinity. These results suggest that LGR4 and RNF43/ZNRF3 form a 2:2 dimer that accommodates bivalent binding of RSPO whereas LGR5 forms a homodimer that does not. Structural models are proposed to illustrate how RSPOs bind to LGR4, RNF43/ZNRF3, and LGR5 in whole cells.
Collapse
Affiliation(s)
- Yukimatsu Toh
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Ling Wu
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Soohyun Park
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Allison Wang
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Jianghua Tu
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Wangsheng Yu
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Mingxin Zuo
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Kendra S Carmon
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA
| | - Qingyun J Liu
- Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Suite 330E, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Wu D, Wang X, Yang X, Gu L, McGeachy MJ, Liu X. Temporary consumption of western diet trains the immune system to reduce future gut inflammation. iScience 2023; 26:106915. [PMID: 37305694 PMCID: PMC10250831 DOI: 10.1016/j.isci.2023.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/23/2023] [Accepted: 05/14/2023] [Indexed: 06/13/2023] Open
Abstract
Urbanization drives the popularity of western diet (WD), which increased burden in metabolic diseases but also in inflammatory diseases. Here, we show continuous WD disrupted the gut barrier, initiating low-grade inflammation and enhancing the colitis response. Nevertheless, transient WD consumption followed by ad libitum normal diet enhanced mucin production and tight junction protein expression in recovered mice. Furthermore, transient WD consumption surprisingly reduced the subsequent inflammatory response in DSS colitis and Citrobacter rodentium-infection induced colitis. The protective effect of WD training was not sex-dependent, and co-housing experiments suggested microbiota changes were not responsible. We identified important roles for cholesterol biosynthesis pathway and macrophages, pointing to innate myeloid training. Together, these data suggest detrimental effects of WD consumption can be reversed on return to a healthier diet. Furthermore, transient WD consumption leads to beneficial immune training, suggesting an evolutionary mechanism to benefit from feasting when abundant food is available.
Collapse
Affiliation(s)
- Dongwen Wu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xiaotong Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xiang Yang
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Lei Gu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Mandy J. McGeachy
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
25
|
Schaaf CR, Polkoff KM, Carter A, Stewart AS, Sheahan B, Freund J, Ginzel J, Snyder JC, Roper J, Piedrahita JA, Gonzalez LM. A LGR5 reporter pig model closely resembles human intestine for improved study of stem cells in disease. FASEB J 2023; 37:e22975. [PMID: 37159340 PMCID: PMC10446885 DOI: 10.1096/fj.202300223r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/12/2023] [Accepted: 05/01/2023] [Indexed: 05/11/2023]
Abstract
Intestinal epithelial stem cells (ISCs) are responsible for intestinal epithelial barrier renewal; thereby, ISCs play a critical role in intestinal pathophysiology research. While transgenic ISC reporter mice are available, advanced translational studies lack a large animal model. This study validates ISC isolation in a new porcine Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5) reporter line and demonstrates the use of these pigs as a novel colorectal cancer (CRC) model. We applied histology, immunofluorescence, fluorescence-activated cell sorting, flow cytometry, gene expression quantification, and 3D organoid cultures to whole tissue and single cells from the duodenum, jejunum, ileum, and colon of LGR5-H2B-GFP and wild-type pigs. Ileum and colon LGR5-H2B-GFP, healthy human, and murine biopsies were compared by mRNA fluorescent in situ hybridization (FISH). To model CRC, adenomatous polyposis coli (APC) mutation was induced by CRISPR/Cas9 editing in porcine LGR5-H2B-GFP colonoids. Crypt-base, green fluorescent protein (GFP) expressing cells co-localized with ISC biomarkers. LGR5-H2B-GFPhi cells had significantly higher LGR5 expression (p < .01) and enteroid forming efficiency (p < .0001) compared with LGR5-H2B-GFPmed/lo/neg cells. Using FISH, similar LGR5, OLFM4, HOPX, LYZ, and SOX9 expression was identified between human and LGR5-H2B-GFP pig crypt-base cells. LGR5-H2B-GFP/APCnull colonoids had cystic growth in WNT/R-spondin-depleted media and significantly upregulated WNT/β-catenin target gene expression (p < .05). LGR5+ ISCs are reproducibly isolated in LGR5-H2B-GFP pigs and used to model CRC in an organoid platform. The known anatomical and physiologic similarities between pig and human, and those shown by crypt-base FISH, underscore the significance of this novel LGR5-H2B-GFP pig to translational ISC research.
Collapse
Affiliation(s)
- Cecilia R. Schaaf
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Kathryn M. Polkoff
- Department of Molecular Biomedical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Amber Carter
- Department of Molecular Biomedical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Amy S. Stewart
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Breanna Sheahan
- Department of Molecular Biomedical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - John Freund
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Joshua Ginzel
- Department of SurgeryDuke UniversityDurhamNorth CarolinaUSA
| | - Joshua C. Snyder
- Department of SurgeryDuke UniversityDurhamNorth CarolinaUSA
- Department of Cell BiologyDuke UniversityDurhamNorth CarolinaUSA
| | - Jatin Roper
- Department of Medicine, Division of GastroenterologyDuke UniversityDurhamNorth CarolinaUSA
- Department of Pharmacology and Cancer BiologyDuke UniversityDurhamNorth CarolinaUSA
| | - Jorge A. Piedrahita
- Department of Molecular Biomedical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| | - Liara M. Gonzalez
- Department of Clinical Sciences, College of Veterinary MedicineNorth Carolina State UniversityRaleighNorth CarolinaUSA
| |
Collapse
|
26
|
Borlongan MC, Wang H. Profiling and targeting cancer stem cell signaling pathways for cancer therapeutics. Front Cell Dev Biol 2023; 11:1125174. [PMID: 37305676 PMCID: PMC10247984 DOI: 10.3389/fcell.2023.1125174] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Tumorigenic cancer stem cells (CSCs) represent a subpopulation of cells within the tumor that express genetic and phenotypic profiles and signaling pathways distinct from the other tumor cells. CSCs have eluded many conventional anti-oncogenic treatments, resulting in metastases and relapses of cancers. Effectively targeting CSCs' unique self-renewal and differentiation properties would be a breakthrough in cancer therapy. A better characterization of the CSCs' unique signaling mechanisms will improve our understanding of the pathology and treatment of cancer. In this paper, we will discuss CSC origin, followed by an in-depth review of CSC-associated signaling pathways. Particular emphasis is given on CSC signaling pathways' ligand-receptor engagement, upstream and downstream mechanisms, and associated genes, and molecules. Signaling pathways associated with regulation of CSC development stand as potential targets of CSC therapy, which include Wnt, TGFβ (transforming growth factor-β)/SMAD, Notch, JAK-STAT (Janus kinase-signal transducers and activators of transcription), Hedgehog (Hh), and vascular endothelial growth factor (VEGF). Lastly, we will also discuss milestone discoveries in CSC-based therapies, including pre-clinical and clinical studies featuring novel CSC signaling pathway cancer therapeutics. This review aims at generating innovative views on CSCs toward a better understanding of cancer pathology and treatment.
Collapse
Affiliation(s)
- Mia C. Borlongan
- Master Program of Pharmaceutical Science College of Graduate Studies, Elk Grove, CA, United States
| | - Hongbin Wang
- Master Program of Pharmaceutical Science College of Graduate Studies, Elk Grove, CA, United States
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, Elk Grove, CA, United States
- Department of Basic Science College of Medicine, California Northstate University, Elk Grove, CA, United States
| |
Collapse
|
27
|
He K, Gan WJ. Wnt/β-Catenin Signaling Pathway in the Development and Progression of Colorectal Cancer. Cancer Manag Res 2023; 15:435-448. [PMID: 37250384 PMCID: PMC10224676 DOI: 10.2147/cmar.s411168] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023] Open
Abstract
The Wnt/β-catenin signaling pathway is a growth control pathway involved in various biological processes as well as the development and progression of cancer. Colorectal cancer (CRC) is one of the most common malignancies in the world. The hyperactivation of Wnt signaling is observed in almost all CRC and plays a crucial role in cancer-related processes such as cancer stem cell (CSC) propagation, angiogenesis, epithelial-mesenchymal transition (EMT), chemoresistance, and metastasis. This review will discuss how the Wnt/β-catenin signaling pathway is involved in the carcinogenesis and progression of CRC and related therapeutic approaches.
Collapse
Affiliation(s)
- Kuang He
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, Jiangsu, People’s Republic of China
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Suzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
28
|
van Kerkhof P, Kralj T, Spanevello F, van Bloois L, Jordens I, van der Vaart J, Jamieson C, Merenda A, Mastrobattista E, Maurice MM. RSPO3 Furin domain-conjugated liposomes for selective drug delivery to LGR5-high cells. J Control Release 2023; 356:72-83. [PMID: 36813038 DOI: 10.1016/j.jconrel.2023.02.025] [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: 09/02/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
The transmembrane receptor LGR5 potentiates Wnt/β-catenin signaling by binding both secreted R-spondin (RSPOs) and the Wnt tumor suppressors RNF43/ZNRF3, directing clearance of RNF43/ZNRF3 from the cell surface. Besides being widely used as a stem cell marker in various tissues, LGR5 is overexpressed in many types of malignancies, including colorectal cancer. Its expression characterizes a subpopulation of cancer cells that play a crucial role in tumor initiation, progression and cancer relapse, known as cancer stem cells (CSCs). For this reason, ongoing efforts are aimed at eradicating LGR5-positive CSCs. Here, we engineered liposomes decorated with different RSPO proteins to specifically detect and target LGR5-positive cells. Using fluorescence-loaded liposomes, we show that conjugation of full-length RSPO1 to the liposomal surface mediates aspecific, LGR5-independent cellular uptake, largely mediated by heparan sulfate proteoglycan binding. By contrast, liposomes decorated only with the Furin (FuFu) domains of RSPO3 are taken up by cells in a highly specific, LGR5-dependent manner. Moreover, encapsulating doxorubicin in FuFuRSPO3 liposomes allowed us to selectively inhibit the growth of LGR5-high cells. Thus, FuFuRSPO3-coated liposomes allow for the selective detection and ablation of LGR5-high cells, providing a potential drug delivery system for LGR5-targeted anti-cancer strategies.
Collapse
Affiliation(s)
- Peter van Kerkhof
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Tomica Kralj
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Francesca Spanevello
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Louis van Bloois
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Ingrid Jordens
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Jelte van der Vaart
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Cara Jamieson
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Alessandra Merenda
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands.
| | - Madelon M Maurice
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
29
|
Zhang X, Li C, Wu Y, Cui P. The research progress of Wnt/β-catenin signaling pathway in colorectal cancer. Clin Res Hepatol Gastroenterol 2023; 47:102086. [PMID: 36657523 DOI: 10.1016/j.clinre.2023.102086] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/05/2022] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
The Wnt/β-catenin signaling pathway is highly conservative. β-catenin is the key molecule in this pathway. The β-catenin target genes regulate cell proliferation and apoptosis. Since Wnt pathway proteins are distributed on the cell membrane, cytoplasm, and nucleus, inhibiting or activating these pathway proteins presents a novel target for cancer treatment via the Wnt signaling pathway. Studies have found that this pathway plays a significant role in the formation and progression of cancers, particularly colorectal cancer. We summarised the activation and inhibition of the Wnt signaling pathway in tumors, its relationship with the microenvironment and crosstalk with other pathways, and the effect of targeting abnormal Wnt signaling in the treatment of colorectal cancer. Here is to review future targeted therapeutics in colorectal cancer research and implementation.
Collapse
Affiliation(s)
- Xueling Zhang
- Department of Internal Medicine, International Medical Services (IMS), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Congcong Li
- Department of Internal Medicine, International Medical Services (IMS), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yue Wu
- Department of Internal Medicine, International Medical Services (IMS), Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peilin Cui
- Department of Internal Medicine, International Medical Services (IMS), Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Gastroenterology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
30
|
Guevara-Garcia A, Soleilhac M, Minc N, Delacour D. Regulation and functions of cell division in the intestinal tissue. Semin Cell Dev Biol 2023:S1084-9521(23)00004-6. [PMID: 36702722 DOI: 10.1016/j.semcdb.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023]
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing epithelial tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs to ensure correct organ formation and functioning. In these processes, proliferation rates and division orientation regulate the speed, timing and direction of tissue expansion but also its proper patterning. Moreover, tissue homeostasis relies on spatio-temporal modulations of daughter cell behavior and arrangement. These aspects are particularly crucial in the intestine, which is one of the most proliferative tissues in adults, making it a very attractive adult organ system to study the role of cell division on epithelial morphogenesis and organ function. Although epithelial cell division has been the subject of intense research for many years in multiple models, it still remains in its infancy in the context of the intestinal tissue. In this review, we focus on the current knowledge on cell division and regulatory mechanisms at play in the intestinal epithelial tissue, as well as their importance in developmental biology and physiopathology.
Collapse
Affiliation(s)
| | - Matis Soleilhac
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Nicolas Minc
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France
| | - Delphine Delacour
- Université de Paris, CNRS, Institut Jacques Monod, F-75006 Paris, France.
| |
Collapse
|
31
|
LRRC superfamily expression in stromal cells predicts the clinical prognosis and platinum resistance of ovarian cancer. BMC Med Genomics 2023; 16:10. [PMID: 36653841 PMCID: PMC9850808 DOI: 10.1186/s12920-023-01435-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Leucine-rich repeat sequence domains are known to mediate protein‒protein interactions. Recently, some studies showed that members of the leucine rich repeat containing (LRRC) protein superfamily may become new targets for the diagnosis and treatment of tumours. However, it is not known whether any of the LRRC superfamily genes is expressed in the stroma of ovarian cancer (OC) and is associated with prognosis. METHODS The clinical data and transcriptional profiles of OC patients from the public databases TCGA (n = 427), GTEx (n = 88) and GEO (GSE40266 and GSE40595) were analysed by R software. A nomogram model was also generated through R. An online public database was used for auxiliary analysis of prognosis, immune infiltration and protein‒protein interaction (PPI) networks. Immunohistochemistry and qPCR were performed to determine the protein and mRNA levels of genes in high-grade serous ovarian cancer (HGSC) tissues of participants and the MRC-5 cell line induced by TGF-β. RESULTS LRRC15 and LRRC32 were identified as differentially expressed genes from the LRRC superfamily by GEO transcriptome analysis. PPI network analysis suggested that they were most enriched in TGF-β signalling. The TCGA-GTEx analysis results showed that only LRRC15 was highly expressed in both cancer-associated fibroblasts (CAFs) and the tumour stroma of OC and was related to clinical prognosis. Based on this, we developed a nomogram model to predict the incidence of adverse outcomes in OC. Moreover, LRRC15 was positively correlated with CAF infiltration and negatively correlated with CD8 + T-cell infiltration. As a single indicator, LRRC15 had the highest accuracy (AUC = 0.920) in predicting the outcome of primary platinum resistance. CONCLUSIONS The LRRC superfamily is related to the TGF-β pathway in the microenvironment of OC. LRRC15, as a stromal biomarker, can predict the clinical prognosis of HGSC and promote the immunosuppressive microenvironment. LRRC15 may be a potential therapeutic target for reversing primary resistance in OC.
Collapse
|
32
|
Imaimatsu K, Uchida A, Hiramatsu R, Kanai Y. Gonadal Sex Differentiation and Ovarian Organogenesis along the Cortical-Medullary Axis in Mammals. Int J Mol Sci 2022; 23:13373. [PMID: 36362161 PMCID: PMC9655463 DOI: 10.3390/ijms232113373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/24/2022] [Accepted: 10/31/2022] [Indexed: 09/20/2023] Open
Abstract
In most mammals, the sex of the gonads is based on the fate of the supporting cell lineages, which arises from the proliferation of coelomic epithelium (CE) that surfaces on the bipotential genital ridge in both XY and XX embryos. Recent genetic studies and single-cell transcriptome analyses in mice have revealed the cellular and molecular events in the two-wave proliferation of the CE that produce the supporting cells. This proliferation contributes to the formation of the primary sex cords in the medullary region of both the testis and the ovary at the early phase of gonadal sex differentiation, as well as to that of the secondary sex cords in the cortical region of the ovary at the perinatal stage. To support gametogenesis, the testis forms seminiferous tubules in the medullary region, whereas the ovary forms follicles mainly in the cortical region. The medullary region in the ovary exhibits morphological and functional diversity among mammalian species that ranges from ovary-like to testis-like characteristics. This review focuses on the mechanism of gonadal sex differentiation along the cortical-medullary axis and compares the features of the cortical and medullary regions of the ovary in mammalian species.
Collapse
Affiliation(s)
- Kenya Imaimatsu
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Aya Uchida
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
- RIKEN BioResouce Research Center, Tsukuba 305-0074, Japan
| | - Ryuji Hiramatsu
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Yoshiakira Kanai
- Department of Veterinary Anatomy, The University of Tokyo, Bunkyo-ku, Tokyo 113-8654, Japan
| |
Collapse
|
33
|
Ding S, Zhang X, Qiu H, Wo J, Zhang F, Na J. Non-cardiomyocytes in the heart in embryo development, health, and disease, a single-cell perspective. Front Cell Dev Biol 2022; 10:873264. [PMID: 36393852 PMCID: PMC9661523 DOI: 10.3389/fcell.2022.873264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
Recent single-cell atlases of the heart gave unprecedented details about the diversity of cell types and states during heart development in health and disease conditions. Beyond a profiling tool, researchers also use single-cell analyses to dissect the mechanism of diseases in animal models. The new knowledge from these studies revealed that beating cardiomyocytes account for less than 50% of the total heart cell population. In contrast, non-cardiomyocytes (NCMs), such as cardiac fibroblasts, endothelial cells, and immune cells, make up the remaining proportion and have indispensable roles in structural support, homeostasis maintenance, and injury repair of the heart. In this review, we categorize the composition and characteristics of NCMs from the latest single-cell studies of the heart in various contexts and compare the findings from both human samples and mouse models. This information will enrich our understanding of the cellular basis of heart development and diseases and provide insights into the potential therapeutic targets in NCMs to repair the heart.
Collapse
Affiliation(s)
- Shuangyuan Ding
- School of Medicine, Tsinghua University, Beijing, China
- Center for Life Sciences, Tsinghua University and Peking University, Beijing, China
- *Correspondence: Shuangyuan Ding, ; Jie Na,
| | - Xingwu Zhang
- School of Medicine, Tsinghua University, Beijing, China
| | - Hui Qiu
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiaoyang Wo
- Center for Life Sciences, Tsinghua University and Peking University, Beijing, China
| | - Fengzhi Zhang
- Central Laboratory, First Hospital of Tsinghua University, Beijing, China
| | - Jie Na
- School of Medicine, Tsinghua University, Beijing, China
- *Correspondence: Shuangyuan Ding, ; Jie Na,
| |
Collapse
|
34
|
Li JY, Gillilland M, Lee AA, Wu X, Zhou SY, Owyang C. Secondary bile acids mediate high-fat diet-induced upregulation of R-spondin 3 and intestinal epithelial proliferation. JCI Insight 2022; 7:e148309. [PMID: 36099053 PMCID: PMC9675439 DOI: 10.1172/jci.insight.148309] [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: 02/04/2021] [Accepted: 08/31/2022] [Indexed: 11/17/2022] Open
Abstract
A high-fat diet (HFD) contributes to the increased incidence of colorectal cancer, but the mechanisms are unclear. We found that R-spondin 3 (Rspo3), a ligand for leucine-rich, repeat-containing GPCR 4 and 5 (LGR4 and LGR5), was the main subtype of R-spondins and was produced by myofibroblasts beneath the crypts in the intestine. HFD upregulated colonic Rspo3, LGR4, LGR5, and β-catenin gene expression in specific pathogen-free rodents, but not in germ-free mice, and the upregulations were prevented by the bile acid (BA) binder cholestyramine or antibiotic treatment, indicating mediation by both BA and gut microbiota. Cholestyramine or antibiotic treatments prevented HFD-induced enrichment of members of the Lachnospiraceae and Rumincoccaceae, which can transform primary BA into secondary BA. Oral administration of deoxycholic acid (DCA), or inoculation of a combination of the BA deconjugator Lactobacillus plantarum and 7α-dehydroxylase-containing Clostridium scindens with an HFD to germ-free mice increased serum DCA and colonic Rspo3 mRNA levels, indicating that formation of secondary BA by gut microbiota is responsible for HFD-induced upregulation of Rspo3. In primary myofibroblasts, DCA increased Rspo3 mRNA via TGR5. Finally, we showed that cholestyramine or conditional deletion of Rspo3 prevented HFD- or DCA-induced intestinal proliferation. We conclude that secondary BA is responsible for HFD-induced upregulation of Rspo3, which, in turn, mediates HFD-induced intestinal epithelial proliferation.
Collapse
|
35
|
Olbertová K, Hrčkulák D, Kříž V, Jesionek W, Kubovčiak J, Ešner M, Kořínek V, Buchtová M. Role of LGR5-positive mesenchymal cells in craniofacial development. Front Cell Dev Biol 2022; 10:810527. [PMID: 36133922 PMCID: PMC9484000 DOI: 10.3389/fcell.2022.810527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 08/03/2022] [Indexed: 11/28/2022] Open
Abstract
Leucine Rich Repeat Containing G Protein-Coupled Receptor 5 (LGR5), a Wnt pathway member, has been previously recognised as a stem cell marker in numerous epithelial tissues. In this study, we used Lgr5-EGFP-CreERT2 mice to analyse the distribution of LGR5-positive cells during craniofacial development. LGR5 expressing cells were primarily located in the mesenchyme adjacent to the craniofacial epithelial structures undergoing folding, such as the nasopharyngeal duct, lingual groove, and vomeronasal organ. To follow the fate of LGR5-positive cells, we performed lineage tracing using an inducible Cre knock-in allele in combination with Rosa26-tdTomato reporter mice. The slight expansion of LGR5-positive cells was found around the vomeronasal organ, in the nasal cavity, and around the epithelium in the lingual groove. However, most LGR5 expressing cells remained in their original location, possibly supporting their signalling function for adjacent epithelium rather than exerting their role as progenitor cells for the craniofacial structures. Moreover, Lgr5 knockout mice displayed distinct defects in LGR5-positive areas, especially in the reduction of the nasopharyngeal duct, the alteration of the palatal shelves shape, abnormal epithelial folding in the lingual groove area, and the disruption of salivary gland development. The latter defect manifested as an atypical number and localisation of the glandular ducts. The gene expression of several Wnt pathway members (Rspo1-3, Axin2) was altered in Lgr5-deficient animals. However, the difference was not found in sorted EGFP-positive cells obtained from Lgr5 +/+ and Lgr5 -/- animals. Expression profiling of LGR5-positive cells revealed the expression of several markers of mesenchymal cells, antagonists, as well as agonists, of Wnt signalling, and molecules associated with the basal membrane. Therefore, LGR5-positive cells in the craniofacial area represent a very specific population of mesenchymal cells adjacent to the epithelium undergoing folding or groove formation. Our results indicate a possible novel role of LGR5 in the regulation of morphogenetic processes during the formation of complex epithelial structures in the craniofacial areas, a role which is not related to the stem cell properties of LGR5-positive cells as was previously defined for various epithelial tissues.
Collapse
Affiliation(s)
- Kristýna Olbertová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Dušan Hrčkulák
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Vítězslav Kříž
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Wojciech Jesionek
- Cellular Imaging Core Facility, Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Milan Ešner
- Cellular Imaging Core Facility, Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czechia
| | - Vladimír Kořínek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics, Czech Academy of Sciences, Prague, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
36
|
Cyr DG, Pinel L. Emerging organoid models to study the epididymis in male reproductive toxicology. Reprod Toxicol 2022; 112:88-99. [PMID: 35810924 DOI: 10.1016/j.reprotox.2022.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/13/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
The importance of the epididymis on sperm maturation and consequently male fertility has been well documented. The pseudostratified epithelium of the epididymis is comprised of multiple cell types, including principal cells, which are the most abundant, and basal cells. The role of basal cells has been unclear and has been a source of discussion in the literature. However, the recent demonstration that these cells are multipotent or adult stem cells has opened new areas of research in epididymal biology. One such avenue is to understand the regulation of these stem cells, and to exploit their properties to develop tools for toxicological studies to elucidate the effects of chemicals on cell differentiation and epididymal function in vitro. Studies in both rat and mouse have shown that purified single epididymal basal cells cultured under 3D conditions can proliferate and differentiate to form organoids, or mini organs. Furthermore, these epididymal basal stem cells can self-renew and differentiate into other epididymal cell types. It is known that during epididymal development, basal cells are derived from undifferentiated columnar cells, which have been reported to share common properties to stem cells. Like basal cells, these undifferentiated columnar cells can also form organoids under 3D culture conditions and can differentiate into basal, principal and clear cells. Organoids derived from either basal cells or columnar cells offer unique models for toxicology studies and represent an exciting and emerging approach to understand the epididymis.
Collapse
Affiliation(s)
- Daniel G Cyr
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada; Department of Obstetrics, Gynecology, and Reproduction, Laval University, Québec, QC, Canada.
| | - Laurie Pinel
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| |
Collapse
|
37
|
Extracellular Vesicles—Oral Therapeutics of the Future. Int J Mol Sci 2022; 23:ijms23147554. [PMID: 35886902 PMCID: PMC9315796 DOI: 10.3390/ijms23147554] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
Considered an artifact just after discovery, the possibility of oral delivery of extracellular vesicles (EVs) and their functional cargos has recently gained much research attention. EVs from various sources, including edible plants, milk, bacteria and mammalian cells, have emerged as a platform for miRNA and drug delivery that seem to induce the expected immune effects locally and in distant tissues after oral administration. Such a possibility greatly expands the clinical applicability of EVs. The present review summarizes research findings that either support or deny the biological/therapeutical activity of orally administered EVs and their role in cross-species and cross-kingdom signaling.
Collapse
|
38
|
Dufresne J, Gregory M, Pinel L, Cyr DG. Differential gene expression and hallmarks of stemness in epithelial cells of the developing rat epididymis. Cell Tissue Res 2022; 389:327-349. [PMID: 35590013 DOI: 10.1007/s00441-022-03634-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/05/2022] [Indexed: 01/07/2023]
Abstract
Epididymal development can be subdivided into three phases: undifferentiated, a period of differentiation, and expansion. The objectives of this study were (1) to assess gene expression profiles in epididymides, (2) predict signaling pathways, and (3) develop a novel 3D cell culture method to assess the regulation of epididymal development in vitro. Microarray analyses indicate that the largest changes in differential gene expression occurred between the 7- to 18-day period, in which 1452 genes were differentially expressed, while 671 differentially expressed genes were noted between days 18 and 28, and there were 560 differentially expressed genes between days 28 and 60. Multiple signaling pathways were predicted at different phases of development. Pathway associations indicated that in epididymides of 7- to 18-day old rats, there was a significant association of regulated genes implicated in stem cells, estrogens, thyroid hormones, and kidney development, while androgen- and estrogen-related pathways were enriched at other phases of development. Organoids were derived from CD49f + columnar cells from 7-day old rats, while no organoids developed from CD49f- cells. Cells cultured in an epididymal basal cell organoid medium versus a commercial kidney differentiation medium supplemented with DHT revealed that irrespective of the culture medium, cells within differentiating organoids expressed p63, AQP9, and V-ATPase after 14 days of culture. The commercial kidney medium resulted in an increase in the number of organoids positive for p63, AQP9, and V-ATPase. Together, these data indicate that columnar cells represent an epididymal stem/progenitor cell population.
Collapse
Affiliation(s)
- Julie Dufresne
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, 245 boul. Des Prairies, Laval, QC, H7V 3B7, Canada
| | - Mary Gregory
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, 245 boul. Des Prairies, Laval, QC, H7V 3B7, Canada
| | - Laurie Pinel
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, 245 boul. Des Prairies, Laval, QC, H7V 3B7, Canada
| | - Daniel G Cyr
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, 245 boul. Des Prairies, Laval, QC, H7V 3B7, Canada. .,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada. .,Department of Obstetrics, Gynecology, and Reproduction, Laval University, Québec, QC, Canada.
| |
Collapse
|
39
|
Han L, Wei X, Liu C, Volpe G, Zhuang Z, Zou X, Wang Z, Pan T, Yuan Y, Zhang X, Fan P, Guo P, Lai Y, Lei Y, Liu X, Yu F, Shangguan S, Lai G, Deng Q, Liu Y, Wu L, Shi Q, Yu H, Huang Y, Cheng M, Xu J, Liu Y, Wang M, Wang C, Zhang Y, Xie D, Yang Y, Yu Y, Zheng H, Wei Y, Huang F, Lei J, Huang W, Zhu Z, Lu H, Wang B, Wei X, Chen F, Yang T, Du W, Chen J, Xu S, An J, Ward C, Wang Z, Pei Z, Wong CW, Liu X, Zhang H, Liu M, Qin B, Schambach A, Isern J, Feng L, Liu Y, Guo X, Liu Z, Sun Q, Maxwell PH, Barker N, Muñoz-Cánoves P, Gu Y, Mulder J, Uhlen M, Tan T, Liu S, Yang H, Wang J, Hou Y, Xu X, Esteban MA, Liu L. Cell transcriptomic atlas of the non-human primate Macaca fascicularis. Nature 2022; 604:723-731. [PMID: 35418686 DOI: 10.1038/s41586-022-04587-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022]
Abstract
Studying tissue composition and function in non-human primates (NHPs) is crucial to understand the nature of our own species. Here we present a large-scale cell transcriptomic atlas that encompasses over 1 million cells from 45 tissues of the adult NHP Macaca fascicularis. This dataset provides a vast annotated resource to study a species phylogenetically close to humans. To demonstrate the utility of the atlas, we have reconstructed the cell-cell interaction networks that drive Wnt signalling across the body, mapped the distribution of receptors and co-receptors for viruses causing human infectious diseases, and intersected our data with human genetic disease orthologues to establish potential clinical associations. Our M. fascicularis cell atlas constitutes an essential reference for future studies in humans and NHPs.
Collapse
Affiliation(s)
- Lei Han
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiaoyu Wei
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chuanyu Liu
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Zhenkun Zhuang
- BGI-Shenzhen, Shenzhen, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xuanxuan Zou
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhifeng Wang
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China
| | - Taotao Pan
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China
| | - Yue Yuan
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Zhang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Peng Fan
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pengcheng Guo
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yiwei Lai
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ying Lei
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Xingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Feng Yu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shuncheng Shangguan
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
| | - Guangyao Lai
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
| | - Qiuting Deng
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Liu
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China
| | - Liang Wu
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Quan Shi
- BGI-Shenzhen, Shenzhen, China.,Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Hao Yu
- BGI-Shenzhen, Shenzhen, China
| | - Yunting Huang
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Mengnan Cheng
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiangshan Xu
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Liu
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Chunqing Wang
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yuanhang Zhang
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Duo Xie
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yunzhi Yang
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yeya Yu
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huiwen Zheng
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanrong Wei
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fubaoqian Huang
- BGI-Shenzhen, Shenzhen, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Junjie Lei
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Waidong Huang
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyong Zhu
- BGI-Shenzhen, Shenzhen, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Haorong Lu
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Bo Wang
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Xiaofeng Wei
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Fengzhen Chen
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Tao Yang
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Wensi Du
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Jing Chen
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Shibo Xu
- Institute for Stem Cells and Neural Regeneration, School of Pharmacy, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Juan An
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,University of Science and Technology of China, Hefei, China
| | - Carl Ward
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zongren Wang
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong Pei
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Xiaolei Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Huafeng Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingyuan Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baoming Qin
- Laboratory of Metabolism and Cell Fate, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Harvard Medical School, MA, Boston, USA
| | - Joan Isern
- Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain
| | - Liqiang Feng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yan Liu
- Institute for Stem Cells and Neural Regeneration, School of Pharmacy, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xiangyu Guo
- Jinan University, Guangzhou, China.,Hubei Topgene Biotechnology Co., Ltd, Wuhan, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Patrick H Maxwell
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nick Barker
- A*STAR Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Pura Muñoz-Cánoves
- Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), ICREA and CIBERNED, Barcelona, Spain
| | - Ying Gu
- BGI-Shenzhen, Shenzhen, China
| | - Jan Mulder
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Mathias Uhlen
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Tao Tan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Shiping Liu
- BGI-Shenzhen, Shenzhen, China.,BGI-Beijing, Beijing, China.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China.,James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, China.,James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Yong Hou
- BGI-Shenzhen, Shenzhen, China. .,BGI-Beijing, Beijing, China. .,Shenzhen Bay Laboratory, Shenzhen, China. .,BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, China. .,BGI-Beijing, Beijing, China. .,BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China. .,Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China.
| | - Miguel A Esteban
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China. .,Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen, China. .,BGI-Beijing, Beijing, China. .,Shenzhen Bay Laboratory, Shenzhen, China. .,BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
40
|
Donowitz M. Has a physiologic function for NHE2 finally been identified? Acta Physiol (Oxf) 2022; 234:e13792. [PMID: 35094506 PMCID: PMC11283681 DOI: 10.1111/apha.13792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Mark Donowitz
- Department of Medicine and Physiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
41
|
Lin X, Gaudino SJ, Jang KK, Bahadur T, Singh A, Banerjee A, Beaupre M, Chu T, Wong HT, Kim CK, Kempen C, Axelrad J, Huang H, Khalid S, Shah V, Eskiocak O, Parks OB, Berisha A, McAleer JP, Good M, Hoshino M, Blumberg R, Bialkowska AB, Gaffen SL, Kolls JK, Yang VW, Beyaz S, Cadwell K, Kumar P. IL-17RA-signaling in Lgr5 + intestinal stem cells induces expression of transcription factor ATOH1 to promote secretory cell lineage commitment. Immunity 2022; 55:237-253.e8. [PMID: 35081371 PMCID: PMC8895883 DOI: 10.1016/j.immuni.2021.12.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/06/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022]
Abstract
The Th17 cell-lineage-defining cytokine IL-17A contributes to host defense and inflammatory disease by coordinating multicellular immune responses. The IL-17 receptor (IL-17RA) is expressed by diverse intestinal cell types, and therapies targeting IL-17A induce adverse intestinal events, suggesting additional tissue-specific functions. Here, we used multiple conditional deletion models to identify a role for IL-17A in secretory epithelial cell differentiation in the gut. Paneth, tuft, goblet, and enteroendocrine cell numbers were dependent on IL-17A-mediated induction of the transcription factor ATOH1 in Lgr5+ intestinal epithelial stem cells. Although dispensable at steady state, IL-17RA signaling in ATOH1+ cells was required to regenerate secretory cells following injury. Finally, IL-17A stimulation of human-derived intestinal organoids that were locked into a cystic immature state induced ATOH1 expression and rescued secretory cell differentiation. Our data suggest that the cross talk between immune cells and stem cells regulates secretory cell lineage commitment and the integrity of the mucosa.
Collapse
Affiliation(s)
- Xun Lin
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Stephen J Gaudino
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Kyung Ku Jang
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Tej Bahadur
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Ankita Singh
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Anirban Banerjee
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Michael Beaupre
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Timothy Chu
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Hoi Tong Wong
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Chang-Kyung Kim
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Cody Kempen
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Jordan Axelrad
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Huakang Huang
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Saba Khalid
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Vyom Shah
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Onur Eskiocak
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Olivia B Parks
- University of Pittsburgh School of Medicine, Medical Scientist Training Program, Pittsburgh, PA 15213, USA
| | - Artan Berisha
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA
| | - Jeremy P McAleer
- Department of Pharmaceutical Science, Marshall University School of Pharmacy, Huntington, WV 25701, USA
| | - Misty Good
- Washington University School of Medicine, Department of Pediatrics, Division of Newborn Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Miko Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Richard Blumberg
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sarah L Gaffen
- Division of Rheumatology and Clinical Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane School of Medicine, New Orleans, LA 70112, USA
| | - Vincent W Yang
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, USA; Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA; Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| | - Pawan Kumar
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
42
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
43
|
Disoma C, Zhou Y, Li S, Peng J, Xia Z. Wnt/β-catenin signaling in colorectal cancer: Is therapeutic targeting even possible? Biochimie 2022; 195:39-53. [DOI: 10.1016/j.biochi.2022.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/03/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
44
|
Bagherani N, Hasić-Mujanović M, Smoller B, Reyes-Barron C, Bergler-Czop B, Miziołek B, Kasumagic-Halilovic E, Sinclair R, Poa JE, Ankad BS, Bagherani N, Sahebnasagh R. Disorders of Hair. ATLAS OF DERMATOLOGY, DERMATOPATHOLOGY AND VENEREOLOGY 2022:669-742. [DOI: 10.1007/978-3-319-53808-2_53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
45
|
Tempest N, Hill CJ, Maclean A, Marston K, Powell SG, Al-Lamee H, Hapangama DK. Novel microarchitecture of human endometrial glands: implications in endometrial regeneration and pathologies. Hum Reprod Update 2021; 28:153-171. [PMID: 34875046 PMCID: PMC8888994 DOI: 10.1093/humupd/dmab039] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/15/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Human endometrium remains a poorly understood tissue of the female reproductive tract. The superficial endometrial functionalis, the site of embryo implantation, is repeatedly shed with menstruation, and the stem cell-rich deeper basalis is postulated to be responsible for the regeneration of the functionalis. Two recent manuscripts have demonstrated the 3D architecture of endometrial glands. These manuscripts have challenged and replaced the prevailing concept that these glands end in blind pouches in the basalis layer that contain stem cells in crypts, as in the intestinal mucosa, providing a new paradigm for endometrial glandular anatomy. This necessitates re-evaluation of the available evidence on human endometrial regeneration in both health and disease in the context of this previously unknown endometrial glandular arrangement. OBJECTIVE AND RATIONALE The aim of this review is to determine if the recently discovered glandular arrangement provides plausible explanations for previously unanswered questions related to human endometrial biology. Specifically, it will focus on re-appraising the theories related to endometrial regeneration, location of stem/progenitor cells and endometrial pathologies in the context of this recently unravelled endometrial glandular organization. SEARCH METHODS An extensive literature search was conducted from inception to April 2021 using multiple databases, including PubMed/Web of Science/EMBASE/Scopus, to select studies using keywords applied to endometrial glandular anatomy and regeneration, and the references included in selected publications were also screened. All relevant publications were included. OUTCOMES The human endometrial glands have a unique and complex architecture; branched basalis glands proceed in a horizontal course adjacent to the myometrium, as opposed to the non-branching, vertically coiled functionalis glands, which run parallel to each other as is observed in intestinal crypts. This complex network of mycelium-like, interconnected basalis glands is demonstrated to contain endometrial epithelial stem cells giving rise to single, non-branching functionalis glands. Several previous studies that have tried to confirm the existence of epithelial stem cells have used methodologies that prevent sampling of the stem cell-rich basalis. More recent findings have provided insight into the efficient regeneration of the human endometrium, which is preferentially evolved in humans and menstruating upper-order primates. WIDER IMPLICATIONS The unique physiological organization of the human endometrial glandular element, its relevance to stem cell activity and scarless endometrial regeneration will inform reproductive biologists and clinicians to direct their future research to determine disease-specific alterations in glandular anatomy in a variety of endometrial pathological conditions.
Collapse
Affiliation(s)
- Nicola Tempest
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK.,Liverpool Women's NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK.,Hewitt Centre for Reproductive Medicine, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Christopher J Hill
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK
| | - Alison Maclean
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK.,Liverpool Women's NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| | - Kathleen Marston
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK
| | - Simon G Powell
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK
| | - Hannan Al-Lamee
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK.,Liverpool Women's NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK.,Hewitt Centre for Reproductive Medicine, Liverpool Women's NHS Foundation Trust, Liverpool, UK
| | - Dharani K Hapangama
- Department of Women's and Children's Health, Centre for Women's Health Research, Institute of Life Course and Medical Sciences, University of Liverpool, Member of Liverpool Health Partners, Liverpool, UK.,Liverpool Women's NHS Foundation Trust, Member of Liverpool Health Partners, Liverpool, UK
| |
Collapse
|
46
|
Miao D, Wang Y, Jia Y, Tong J, Jiang S, Liu L. ZRANB1 enhances stem-cell-like features and accelerates tumor progression by regulating Sox9-mediated USP22/Wnt/β-catenin pathway in colorectal cancer. Cell Signal 2021; 90:110200. [PMID: 34798260 DOI: 10.1016/j.cellsig.2021.110200] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/08/2021] [Accepted: 11/13/2021] [Indexed: 12/18/2022]
Abstract
The pathogenesis of colorectal cancer (CRC) is a multistep process characterized by the accumulation of gene mutations and epigenetic alterations. Tumor necrosis factor receptor-associated factor-binding protein domain (ZRANB1) is a deubiquitinase that mediates tumor growth and metastasis by deubiquitinating target proteins. In this study, we examined the regulatory effects of ZRANB1 on the maintenance of cancer stem cell (CSC) properties and tumor growth in CRC. Human CRC tissue samples and matched normal tissues were collected for the analysis of ZRANB1 expression. ZRANB1 was upregulated in CRC human tissues and cell lines, and its expression was positively correlated with advanced tumor stage and poor survival of CRC patients. The overexpression of ZRANB1 also induced the expression of CSC markers in CRC cells. Then, a xenograft model was established by inoculating BALB/c mice with CRC cells. The upregulation of ZRANB1 promoted tumorigenesis in vivo. Sox9 is a transcription factor that acts as an oncogene in human cancers. ZRANB1 increased the stability of Sox9 in CRC cells by decelerating its ubiquitination. Further analysis revealed that Sox9 regulated the transcription activity of USP22 by binding to its promoter. Moreover, ZRANB1 enhances stem-cell-like features of CRC cells and activated the Wnt/β-catenin pathway through USP22. Our results highlighted the role of ZRANB1 as a molecular target for CRC treatment, which may contribute to the development of novel therapies with better efficacy.
Collapse
Affiliation(s)
- Dazhuang Miao
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Yan Wang
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Yunhe Jia
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China.
| | - Jinxue Tong
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China.
| | - Shixiong Jiang
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Lixiu Liu
- Colorectal Cancer Surgical Ward 2, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
47
|
Modeling Intestinal Stem Cell Function with Organoids. Int J Mol Sci 2021; 22:ijms222010912. [PMID: 34681571 PMCID: PMC8535974 DOI: 10.3390/ijms222010912] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/11/2022] Open
Abstract
Intestinal epithelial cells (IECs) are crucial for the digestive process and nutrient absorption. The intestinal epithelium is composed of the different cell types of the small intestine (mainly, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, and tuft cells). The small intestine is characterized by the presence of crypt-villus units that are in a state of homeostatic cell turnover. Organoid technology enables an efficient expansion of intestinal epithelial tissue in vitro. Thus, organoids hold great promise for use in medical research and in the development of new treatments. At present, the cholinergic system involved in IECs and intestinal stem cells (ISCs) are attracting a great deal of attention. Thus, understanding the biological processes triggered by epithelial cholinergic activation by acetylcholine (ACh), which is produced and released from neuronal and/or non-neuronal tissue, is of key importance. Cholinergic signaling via ACh receptors plays a pivotal role in IEC growth and differentiation. Here, we discuss current views on neuronal innervation and non-neuronal control of the small intestinal crypts and their impact on ISC proliferation, differentiation, and maintenance. Since technology using intestinal organoid culture systems is advancing, we also outline an organoid-based organ replacement approach for intestinal diseases.
Collapse
|
48
|
Cui J, Toh Y, Park S, Yu W, Tu J, Wu L, Li L, Jacob J, Pan S, Carmon KS, Liu QJ. Drug Conjugates of Antagonistic R-Spondin 4 Mutant for Simultaneous Targeting of Leucine-Rich Repeat-Containing G Protein-Coupled Receptors 4/5/6 for Cancer Treatment. J Med Chem 2021; 64:12572-12581. [PMID: 34406767 DOI: 10.1021/acs.jmedchem.1c00395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
LGR4-6 (leucine-rich repeat-containing G-protein-coupled receptors 4, 5, and 6) are three related receptors with an upregulated expression in gastrointestinal cancers to various extents, and LGR5 is enriched in cancer stem cells. Antibody-drug conjugates (ADCs) targeting LGR5 showed a robust antitumor effect in vivo but could not eradicate tumors due to plasticity of LGR5-positive cancer cells. As LGR5-negative cancer cells often express LGR4 or LGR6 or both, we reasoned that simultaneous targeting of all three LGRs may provide a more effective approach. R-spondins (RSPOs) bind to LGR4-6 with high affinity and potentiate Wnt signaling. We identified an RSPO4 furin domain mutant (Q65R) that retains potent LGR binding but no longer potentiates Wnt signaling. Drug conjugates of a peptibody comprising the RSPO4 mutant and IgG1-Fc showed potent cytotoxic effects on cancer cell lines expressing any LGR in vitro and suppressed tumor growth in vivo without inducing intestinal enlargement or other adverse effects.
Collapse
Affiliation(s)
- Jie Cui
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Yukimatsu Toh
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Soohyun Park
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Wangsheng Yu
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Jianghua Tu
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Ling Wu
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Li Li
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Joan Jacob
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Sheng Pan
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Kendra S Carmon
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| | - Qingyun J Liu
- Center for Translational Cancer Research, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Pressler St., Houston, Texas 77030, United States
| |
Collapse
|
49
|
Bittenglova K, Habart D, Saudek F, Koblas T. The Potential of Pancreatic Organoids for Diabetes Research and Therapy. Islets 2021; 13:85-105. [PMID: 34523383 PMCID: PMC8528407 DOI: 10.1080/19382014.2021.1941555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/04/2021] [Indexed: 10/20/2022] Open
Abstract
The success of clinical transplantation of pancreas or isolated pancreatic islets supports the concept of cell-based cure for diabetes. One limitation is the shortage of cadaver human pancreata. The demand-supply gap could potentially be bridged by harnessing the self-renewal capacity of stem cells. Pluripotent stem cells and adult pancreatic stem cells have been explored as possible cell sources. Recently, a system for long-term culture of proposed adult pancreatic stem cells in a form of organoids was developed. Generated organoids partially mimic the architecture and cell-type composition of pancreatic tissue. Here, we review the attempts over the past decade, to utilize the organoid cell culture principles in order to identify, expand, and differentiate the adult pancreatic stem cells from different compartments of mouse and human pancreata. The development of the culture conditions, effects of specific growth factors and small molecules is discussed. The potential utility of the adult pancreatic stem cells is considered in the context of other cell sources.
Collapse
Affiliation(s)
- Katerina Bittenglova
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Habart
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Frantisek Saudek
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomas Koblas
- Department of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
50
|
Koyama S, Kondo K, Ueha R, Kashiwadani H, Heinbockel T. Possible Use of Phytochemicals for Recovery from COVID-19-Induced Anosmia and Ageusia. Int J Mol Sci 2021; 22:8912. [PMID: 34445619 PMCID: PMC8396277 DOI: 10.3390/ijms22168912] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022] Open
Abstract
The year 2020 became the year of the outbreak of coronavirus, SARS-CoV-2, which escalated into a worldwide pandemic and continued into 2021. One of the unique symptoms of the SARS-CoV-2 disease, COVID-19, is the loss of chemical senses, i.e., smell and taste. Smell training is one of the methods used in facilitating recovery of the olfactory sense, and it uses essential oils of lemon, rose, clove, and eucalyptus. These essential oils were not selected based on their chemical constituents. Although scientific studies have shown that they improve recovery, there may be better combinations for facilitating recovery. Many phytochemicals have bioactive properties with anti-inflammatory and anti-viral effects. In this review, we describe the chemical compounds with anti- inflammatory and anti-viral effects, and we list the plants that contain these chemical compounds. We expand the review from terpenes to the less volatile flavonoids in order to propose a combination of essential oils and diets that can be used to develop a new taste training method, as there has been no taste training so far. Finally, we discuss the possible use of these in clinical settings.
Collapse
Affiliation(s)
- Sachiko Koyama
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Kenji Kondo
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan;
| | - Rumi Ueha
- Department of Otolaryngology, Faculty of Medicine, The University of Tokyo, Tokyo 113-8655, Japan;
- Swallowing Center, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Hideki Kashiwadani
- Department of Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan;
| | - Thomas Heinbockel
- Department of Anatomy, College of Medicine, Howard University, Washington, DC 20059, USA
| |
Collapse
|