Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 181(2): 219-222, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32302564

ABSTRACT

Mounting evidence indicates that the nervous system plays a central role in cancer pathogenesis. In turn, cancers and cancer therapies can alter nervous system form and function. This Commentary seeks to describe the burgeoning field of "cancer neuroscience" and encourage multidisciplinary collaboration for the study of cancer-nervous system interactions.


Subject(s)
Neoplasms/metabolism , Nervous System/metabolism , Humans , Neurosciences
2.
Development ; 145(21)2018 11 05.
Article in English | MEDLINE | ID: mdl-30305288

ABSTRACT

The ductal system of the salivary gland has long been postulated to be resistant to radiation-induced damage, a common side effect incurred by head and neck cancer patients receiving radiotherapy. Yet, whether the ducts are capable of regenerating after genotoxic injury, or whether damage to ductal cells induces lineage plasticity, as has been reported in other organ systems, remains unknown. Here, using the murine salivary gland, we show that two ductal progenitor populations, marked exclusively by KRT14 and KIT, maintain non-overlapping ductal compartments after radiation exposure but do so through distinct cellular mechanisms. KRT14+ progenitor cells are fast-cycling cells that proliferate in response to radiation-induced damage in a sustained manner and divide asymmetrically to produce differentiated cells of the larger granulated ducts. Conversely, KIT+ intercalated duct cells are long-lived progenitors for the intercalated ducts that undergo few cell divisions either during homeostasis or after gamma radiation, thus maintaining ductal architecture with slow rates of cell turnover. Together, these data illustrate the regenerative capacity of the salivary ducts and highlight the heterogeneity in the damage responses used by salivary progenitor cells to maintain tissue architecture.


Subject(s)
Radiation Injuries/therapy , Salivary Ducts/pathology , Salivary Ducts/radiation effects , Stem Cell Transplantation , Stem Cells/cytology , Acinar Cells/metabolism , Animals , Animals, Newborn , Asymmetric Cell Division , Cell Lineage , Cell Proliferation , Epithelial Cells/metabolism , Female , Humans , Keratin-14/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Proto-Oncogene Proteins c-kit/metabolism , Radiation Injuries/pathology , Salivary Ducts/metabolism , Submandibular Gland/metabolism , Submandibular Gland/pathology , Submandibular Gland/radiation effects
3.
Proc Natl Acad Sci U S A ; 115(24): 6279-6284, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29794221

ABSTRACT

Xerostomia (dry mouth) is the most common side effect of radiation therapy in patients with head and neck cancer and causes difficulty speaking and swallowing. Since aldehyde dehydrogenase 3A1 (ALDH3A1) is highly expressed in mouse salivary stem/progenitor cells (SSPCs), we sought to determine the role of ALDH3A1 in SSPCs using genetic loss-of-function and pharmacologic gain-of-function studies. Using DarkZone dye to measure intracellular aldehydes, we observed higher aldehyde accumulation in irradiated Aldh3a1-/- adult murine salisphere cells and in situ in whole murine embryonic salivary glands enriched in SSPCs compared with wild-type glands. To identify a safe ALDH3A1 activator for potential clinical testing, we screened a traditional Chinese medicine library and isolated d-limonene, commonly used as a food-flavoring agent, as a single constituent activator. ALDH3A1 activation by d-limonene significantly reduced aldehyde accumulation in SSPCs and whole embryonic glands, increased sphere-forming ability, decreased apoptosis, and improved submandibular gland structure and function in vivo after radiation. A phase 0 study in patients with salivary gland tumors showed effective delivery of d-limonene into human salivary glands following daily oral dosing. Given its safety and bioavailability, d-limonene may be a good clinical candidate for mitigating xerostomia in patients with head and neck cancer receiving radiation therapy.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Aldehydes/metabolism , Cyclohexenes/pharmacology , Radiotherapy/adverse effects , Salivary Glands/metabolism , Terpenes/pharmacology , Xerostomia/metabolism , Animals , Apoptosis/drug effects , Female , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Limonene , Medicine, Chinese Traditional/methods , Mice , Mice, Inbred C57BL , Protective Agents/pharmacology , Salivary Glands/drug effects , Salivary Glands/radiation effects , Stem Cells/drug effects , Stem Cells/metabolism , Submandibular Gland/drug effects , Submandibular Gland/metabolism , Xerostomia/drug therapy
4.
Development ; 144(13): 2517-2528, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28576768

ABSTRACT

The tear-producing lacrimal gland is a tubular organ that protects and lubricates the ocular surface. The lacrimal gland possesses many features that make it an excellent model in which to investigate tubulogenesis, but the cell types and lineage relationships that drive lacrimal gland formation are unclear. Using single-cell sequencing and other molecular tools, we reveal novel cell identities and epithelial lineage dynamics that underlie lacrimal gland development. We show that the lacrimal gland from its earliest developmental stages is composed of multiple subpopulations of immune, epithelial and mesenchymal cell lineages. The epithelial lineage exhibits the most substantial cellular changes, transitioning through a series of unique transcriptional states to become terminally differentiated acinar, ductal and myoepithelial cells. Furthermore, lineage tracing in postnatal and adult glands provides the first direct evidence of unipotent KRT5+ epithelial cells in the lacrimal gland. Finally, we show conservation of developmental markers between the developing mouse and human lacrimal gland, supporting the use of mice to understand human development. Together, our data reveal crucial features of lacrimal gland development that have broad implications for understanding epithelial organogenesis.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Lacrimal Apparatus/cytology , Lacrimal Apparatus/embryology , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Biomarkers/metabolism , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Mice , Phenotype , Sequence Analysis, RNA , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism
5.
J Autoimmun ; 114: 102500, 2020 11.
Article in English | MEDLINE | ID: mdl-32565048

ABSTRACT

Autoimmune-mediated dry eye disease is a pathological feature of multiple disorders including Sjögren's syndrome, lupus and rheumatoid arthritis that has a life-long, detrimental impact on vision and overall quality of life. Although late stage disease outcomes such as epithelial barrier dysfunction, reduced corneal innervation and chronic inflammation have been well characterized in both human patients and mouse models, there is little to no understanding of early pathological processes. Moreover, the mechanisms underlying the loss of cornea homeostasis and disease progression are unknown. Here, we utilize the autoimmune regulatory (Aire)-deficient mouse model of autoimmune-mediated dry eye disease in combination with genome wide transcriptomics, high-resolution imaging and atomic force microscopy to reveal a potential extracellular matrix (ECM)-biomechanical-based mechanism driving cellular and morphological changes at early disease onset. Early disease in the Aire-deficient mouse model is associated with a mild reduction in tear production and moderate immune cell infiltration, allowing for interrogation of cellular, molecular and biomechanical changes largely independent of chronic inflammation. Using these tools, we demonstrate for the first time that the emergence of autoimmune-mediated dry eye disease is associated with an alteration in the biomechanical properties of the cornea. We reveal a dramatic disruption of the synthesis and organization of the extracellular matrix as well as degradation of the epithelial basement membrane during early disease. Notably, we provide evidence that the nerve supply to the cornea is severely reduced at early disease stages and that this is independent of basement membrane destruction or significant immune cell infiltration. Furthermore, diseased corneas display spatial heterogeneity in mechanical, structural and compositional changes, with the limbal compartment often exhibiting the opposite response compared to the central cornea. Despite these differences, however, epithelial hyperplasia is apparent in both compartments, possibly driven by increased activation of IL-1R1 and YAP signaling pathways. Thus, we reveal novel perturbations in corneal biomechanics, matrix organization and cell behavior during the early phase of dry eye that may underlie disease development and progression, presenting new potential targets for therapeutic intervention.


Subject(s)
Autoimmunity , Biomechanical Phenomena , Cornea/immunology , Cornea/pathology , Dry Eye Syndromes/diagnosis , Dry Eye Syndromes/etiology , Animals , Autoimmune Diseases/diagnosis , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Cornea/metabolism , Disease Models, Animal , Dry Eye Syndromes/metabolism , Extracellular Matrix , Humans , Mice , Mice, Knockout , Severity of Illness Index
6.
Genesis ; 57(1): e23282, 2019 01.
Article in English | MEDLINE | ID: mdl-30628162

ABSTRACT

Organs and structures of the vertebrate head perform a plethora of tasks including visualization, digestion, vocalization/communication, auditory functions, and respiration in response to neuronal input. This input is primarily derived from afferent and efferent fibers of the cranial nerves (sensory and motor respectively) and efferent fibers of the cervical sympathetic trunk. Despite their essential contribution to the function and integration of processes necessary for survival, how organ innervation is established remains poorly understood. Furthermore, while it has been appreciated for some time that innervation of organs by cranial nerves is regulated in part by secreted factors and cell surface ligands expressed by those organs, whether nerves also regulate the development of facial organs is only beginning to be elucidated. This review will provide an overview of cranial nerve development in relation to the organs they innervate, and outline their known contributions to craniofacial development, thereby providing insight into how nerves may shape the organs they innervate during development. Throughout, the interaction between different cell and tissue types will be highlighted.


Subject(s)
Cranial Nerves/embryology , Morphogenesis , Neural Crest/embryology , Animals , Humans , Skull/embryology
7.
Genesis ; 56(5): e23211, 2018 05.
Article in English | MEDLINE | ID: mdl-29663717

ABSTRACT

Salivary glands are responsible for maintaining the health of the oral cavity and are routinely damaged by therapeutic radiation for head and neck cancer as well as by autoimmune diseases such as Sjögren's syndrome. Regenerative approaches based on the reactivation of endogenous stem cells or the transplant of exogenous stem cells hold substantial promise in restoring the structure and function of these organs to improve patient quality of life. However, these approaches have been hampered by a lack of knowledge on the identity of salivary stem cell populations and their regulators. In this review we discuss our current knowledge on salivary stem cells and their regulators during organ development, homeostasis and regeneration. As increasing evidence in other systems suggests that progenitor cells may be a source of cancer, we also review whether these same salivary stem cells may also be cancer initiating cells.


Subject(s)
Adult Stem Cells/physiology , Salivary Glands/cytology , Salivary Glands/metabolism , Animals , Cell Differentiation/physiology , Humans , Neoplasms/metabolism , Neoplasms/therapy , Regeneration/physiology
8.
Dev Biol ; 427(1): 12-20, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28511845

ABSTRACT

The tear film protects the terrestrial animal's ocular surface and the lacrimal gland provides important aqueous secretions necessary for its maintenance. Despite the importance of the lacrimal gland in ocular health, molecular aspects of its development remain poorly understood. We have identified a noncoding RNA (miR-205) as an important gene for lacrimal gland development. Mice lacking miR-205 fail to properly develop lacrimal glands, establishing this noncoding RNA as a key regulator of lacrimal gland development. Specifically, more than half of knockout lacrimal glands never initiated, suggesting a critical role of miR-205 at the earliest stages of lacrimal gland development. RNA-seq analysis uncovered several up-regulated miR-205 targets that may interfere with signaling to impair lacrimal gland initiation. Supporting this data, combinatorial epistatic deletion of Fgf10, the driver of lacrimal gland initiation, and miR-205 in mice exacerbates the lacrimal gland phenotype. We develop a molecular rheostat model where miR-205 modulates signaling pathways related to Fgf10 in order to regulate glandular development. These data show that a single microRNA is a key regulator for early lacrimal gland development in mice and highlights the important role of microRNAs during organogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Lacrimal Apparatus/metabolism , MicroRNAs/genetics , Organogenesis/genetics , Animals , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Lacrimal Apparatus/embryology , Lacrimal Apparatus/growth & development , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA/methods , Signal Transduction/genetics
9.
Cells ; 12(10)2023 05 21.
Article in English | MEDLINE | ID: mdl-37408269

ABSTRACT

The lacrimal gland (LG) secretes aqueous tears. Previous studies have provided insights into the cell lineage relationships during tissue morphogenesis. However, little is known about the cell types composing the adult LG and their progenitors. Using scRNAseq, we established the first comprehensive cell atlas of the adult mouse LG to investigate the cell hierarchy, its secretory repertoire, and the sex differences. Our analysis uncovered the complexity of the stromal landscape. Epithelium subclustering revealed myoepithelial cells, acinar subsets, and two novel acinar subpopulations: Tfrchi and Car6hi cells. The ductal compartment contained Wfdc2+ multilayered ducts and an Ltf+ cluster formed by luminal and intercalated duct cells. Kit+ progenitors were identified as: Krt14+ basal ductal cells, Aldh1a1+ cells of Ltf+ ducts, and Sox10+ cells of the Car6hi acinar and Ltf+ epithelial clusters. Lineage tracing experiments revealed that the Sox10+ adult populations contribute to the myoepithelial, acinar, and ductal lineages. Using scRNAseq data, we found that the postnatally developing LG epithelium harbored key features of putative adult progenitors. Finally, we showed that acinar cells produce most of the sex-biased lipocalins and secretoglobins detected in mouse tears. Our study provides a wealth of new data on LG maintenance and identifies the cellular origin of sex-biased tear components.


Subject(s)
Lacrimal Apparatus , Animals , Female , Male , Mice , Lacrimal Apparatus/metabolism , Transcriptome , Epithelium/metabolism , Epithelial Cells/metabolism , Stem Cells/metabolism , WAP Four-Disulfide Core Domain Protein 2/metabolism
10.
Cell Rep ; 40(9): 111307, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36044852

ABSTRACT

Corneal architecture is essential for vision and is greatly perturbed by the absence of tears due to the highly prevalent disorder dry eye. With no regenerative therapies available, pathological alterations of the ocular surface in response to dryness, including persistent epithelial defects and poor wound healing, result in lifelong morbidity. Here, using a mouse model of aqueous-deficient dry eye, we reveal that topical application of the synthetic tear protein Lacripep reverses the pathological outcomes of dry eye through restoring the extensive network of corneal nerves that are essential for tear secretion, barrier function, epithelial homeostasis, and wound healing. Intriguingly, the restorative effects of Lacripep occur despite extensive immune cell infiltration, suggesting tissue reinnervation and regeneration can be achieved under chronic inflammatory conditions. In summary, our data highlight Lacripep as a first-in-class regenerative therapy for returning the cornea to a near homeostatic state in individuals who suffer from dry eye.


Subject(s)
Dry Eye Syndromes , Tears , Cornea/metabolism , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/pathology , Dry Eye Syndromes/therapy , Humans , Nerve Regeneration
11.
Dev Cell ; 57(22): 2550-2565.e5, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36413949

ABSTRACT

Acinar cells are the principal secretory units of multiple exocrine organs. A single-cell, layered, lumenized acinus forms from a large cohort of epithelial progenitors that must initiate and coordinate three cellular programs of acinar specification, namely, lineage progression, secretion, and polarization. Despite this well-known outcome, the mechanism(s) that regulate these complex programs are unknown. Here, we demonstrate that neuronal-epithelial cross-talk drives acinar specification through neuregulin (NRG1)-ERBB3-mTORC2 signaling. Using single-cell and global RNA sequencing of developing murine salivary glands, we identified NRG1-ERBB3 to precisely overlap with acinar specification during gland development. Genetic deletion of Erbb3 prevented cell lineage progression and the establishment of lumenized, secretory acini. Conversely, NRG1 treatment of isolated epithelia was sufficient to recapitulate the development of secretory acini. Mechanistically, we found that NRG1-ERBB3 regulates each developmental program through an mTORC2 signaling pathway. Thus, we reveal that a neuronal-epithelial (NRG1/ERBB3/mTORC2) mechanism orchestrates the creation of functional acini.


Subject(s)
Neuregulins , Signal Transduction , Humans , Mice , Animals , Mechanistic Target of Rapamycin Complex 2 , Acinar Cells , Biological Transport , Neuregulin-1 , Receptor, ErbB-3
12.
Sci Adv ; 8(51): eadc8753, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36542703

ABSTRACT

Salivary gland acinar cells are severely depleted after radiotherapy for head and neck cancer, leading to loss of saliva and extensive oro-digestive complications. With no regenerative therapies available, organ dysfunction is irreversible. Here, using the adult murine system, we demonstrate that radiation-damaged salivary glands can be functionally regenerated via sustained delivery of the neurogenic muscarinic receptor agonist cevimeline. We show that endogenous gland repair coincides with increased nerve activity and acinar cell division that is limited to the first week after radiation, with extensive acinar cell degeneration, dysfunction, and cholinergic denervation occurring thereafter. However, we found that mimicking cholinergic muscarinic input via sustained local delivery of a cevimeline-alginate hydrogel was sufficient to regenerate innervated acini and retain physiological saliva secretion at nonirradiated levels over the long term (>3 months). Thus, we reveal a previously unknown regenerative approach for restoring epithelial organ structure and function that has extensive implications for human patients.

13.
Int Forum Allergy Rhinol ; 11(10): 1443-1451, 2021 10.
Article in English | MEDLINE | ID: mdl-33956392

ABSTRACT

BACKGROUND: Chronic rhinosinusitis (CRS) is characterized by significant accumulation and thickening of mucus in the sinonasal cavities. One contributor of aberrant mucus production and impaired mucociliary clearance (MCC) is altered function of the sinonasal submucosal glands (SMGs), yet contributions of SMGs to upper airway disease initiation and progression remain unknown. The objective of this study was to characterize the morphology and secretory cell identities of the nasal septum SMGs in both healthy and CRS adults. METHODS: Biopsies from adult participants with CRS without nasal polyps (CRSsNP, n = 4), CRS with nasal polyps (CRSwNP, n = 8), and non-CRS controls (n = 14) were collected from the posterior septum. Glandular morphology and mucus markers were investigated using histological techniques and high-resolution confocal microscopy. RESULTS: Analysis revealed a significant decrease in gland density in the posterior septum of CRSsNP (28% ± 6.15%) and CRSwNP (23% ± 3.09%) compared to control participants (53% ± 1.59%, p < 0.0001). Further analysis of the CRS SMG secretory function revealed an overall decrease in Mucin 5B+ gland mucus being produced. Dilated and cystic ductal structures filled with inspissated mucus were also common to CRS glands. CONCLUSION: Here, we describe a significant alteration in SMG structure and function in the adult CRS posterior septum suggesting reduced gland contribution to MCC. The SMGs of both the nose and sinuses may represent targets for future therapeutic approaches.


Subject(s)
Nasal Polyps , Rhinitis , Sinusitis , Chronic Disease , Humans , Mucins , Nasal Mucosa/pathology , Nasal Polyps/pathology , Rhinitis/pathology , Sinusitis/pathology
14.
Biochemistry ; 49(26): 5524-32, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20507176

ABSTRACT

Perlecan is a large multidomain proteoglycan that is essential for normal cartilage development. In this study, perlecan was localized in the pericellular matrix of hypertrophic chondrocytes in developing human cartilage rudiments. Perlecan immunopurified from medium conditioned by cultured human fetal chondrocytes was found to be substituted with heparan sulfate (HS), chondroitin sulfate (CS), and keratan sulfate (KS). Ligand and carbohydrate engagement (LACE) assays demonstrated that immunopurified chondrocyte-derived perlecan formed HS-dependent ternary complexes with fibroblast growth factor (FGF) 2 and either FGF receptors (FGFRs) 1 or 3; however, these complexes were not biologically active in the BaF32 cell system. Chondrocyte-derived perlecan also formed HS-dependent ternary complexes with FGF18 and FGFR3. The proliferation of BaF32 cells expressing FGFR3 was promoted by chondrocyte-derived perlecan in the presence of FGF18, and this activity was reduced by digestion of the HS with either heparinase III or mammalian heparanase. These data suggest that FGF2 and -18 bind to discrete structures on the HS chains attached to chondrocyte-derived perlecan which modulate the growth factor activities. The presence and activity of mammalian heparanase may be important in the turnover of HS and subsequent signaling required for the establishment and maintenance of functional osteo-chondral junctions in long bone growth.


Subject(s)
Chondrocytes/chemistry , Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Signal Transduction/physiology , Bone Development , Cells, Cultured , Culture Media, Conditioned/chemistry , Fibroblast Growth Factor 2/metabolism , Humans , Protein Binding
15.
Cell Rep ; 33(7): 108402, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33207190

ABSTRACT

Salivary proteins are essential for maintaining health in the oral cavity and proximal digestive tract, and they serve as potential diagnostic markers for monitoring human health and disease. However, their precise organ origins remain unclear. Through transcriptomic analysis of major adult and fetal salivary glands and integration with the saliva proteome, the blood plasma proteome, and transcriptomes of 28+ organs, we link human saliva proteins to their source, identify salivary-gland-specific genes, and uncover fetal- and adult-specific gene repertoires. Our results also provide insights into the degree of gene retention during gland maturation and suggest that functional diversity among adult gland types is driven by specific dosage combinations of hundreds of transcriptional regulators rather than by a few gland-specific factors. Finally, we demonstrate the heterogeneity of the human acinar cell lineage. Our results pave the way for future investigations into glandular biology and pathology, as well as saliva's use as a diagnostic fluid.


Subject(s)
Saliva/chemistry , Saliva/metabolism , Salivary Glands/metabolism , Adult , Aged , Female , Fetus , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Humans , Male , Middle Aged , Mouth/metabolism , Proteome/metabolism , Salivary Glands/physiology , Salivary Proteins and Peptides/metabolism , Structure-Activity Relationship , Transcriptome/genetics
16.
Nat Commun ; 9(1): 3922, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254276

ABSTRACT

Organogenesis requires the complex interactions of multiple cell lineages that coordinate their expansion, differentiation, and maturation over time. Here, we profile the cell types within the epithelial and mesenchymal compartments of the murine pancreas across developmental time using a combination of single-cell RNA sequencing, immunofluorescence, in situ hybridization, and genetic lineage tracing. We identify previously underappreciated cellular heterogeneity of the developing mesenchyme and reconstruct potential lineage relationships among the pancreatic mesothelium and mesenchymal cell types. Within the epithelium, we find a previously undescribed endocrine progenitor population, as well as an analogous population in both human fetal tissue and human embryonic stem cells differentiating toward a pancreatic beta cell fate. Further, we identify candidate transcriptional regulators along the differentiation trajectory of this population toward the alpha or beta cell lineages. This work establishes a roadmap of pancreatic development and demonstrates the broad utility of this approach for understanding lineage dynamics in developing organs.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation, Developmental , Pancreas/metabolism , Single-Cell Analysis/methods , Animals , Cell Differentiation/genetics , Cell Line , Epithelium/embryology , Epithelium/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , In Situ Hybridization , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Pancreas/cytology , Pancreas/embryology , Reverse Transcriptase Polymerase Chain Reaction
17.
EMBO Mol Med ; 10(3)2018 03.
Article in English | MEDLINE | ID: mdl-29335337

ABSTRACT

Salivary gland acinar cells are routinely destroyed during radiation treatment for head and neck cancer that results in a lifetime of hyposalivation and co-morbidities. A potential regenerative strategy for replacing injured tissue is the reactivation of endogenous stem cells by targeted therapeutics. However, the identity of these cells, whether they are capable of regenerating the tissue, and the mechanisms by which they are regulated are unknown. Using in vivo and ex vivo models, in combination with genetic lineage tracing and human tissue, we discover a SOX2+ stem cell population essential to acinar cell maintenance that is capable of replenishing acini after radiation. Furthermore, we show that acinar cell replacement is nerve dependent and that addition of a muscarinic mimetic is sufficient to drive regeneration. Moreover, we show that SOX2 is diminished in irradiated human salivary gland, along with parasympathetic nerves, suggesting that tissue degeneration is due to loss of progenitors and their regulators. Thus, we establish a new paradigm that salivary glands can regenerate after genotoxic shock and do so through a SOX2 nerve-dependent mechanism.


Subject(s)
Radiation Injuries/pathology , Radiation Injuries/physiopathology , Regeneration , SOXB1 Transcription Factors/metabolism , Salivary Glands/pathology , Salivary Glands/physiopathology , Acetylcholine/metabolism , Acinar Cells/metabolism , Acinar Cells/radiation effects , Adult , Aged , Animals , Cell Lineage/radiation effects , Cell Proliferation/radiation effects , Chorda Tympani Nerve/pathology , Chorda Tympani Nerve/radiation effects , Female , Homeostasis , Humans , Male , Mice, Inbred C57BL , Middle Aged , Radiation Injuries/metabolism , Radiation, Ionizing , Receptors, Muscarinic/metabolism , Salivary Glands/radiation effects , Signal Transduction , Stem Cells/metabolism , Stem Cells/radiation effects
18.
PLoS One ; 12(9): e0184916, 2017.
Article in English | MEDLINE | ID: mdl-28926640

ABSTRACT

Sjögren's syndrome (SS) is a chronic, autoimmune exocrinopathy that leads to severe dryness of the mouth and eyes. Exocrine function is highly regulated by neuronal mechanisms but little is known about the link between chronic inflammation, innervation and altered exocrine function in the diseased eyes and exocrine glands of SS patients. To gain a better understanding of neuronal regulation in the immunopathogenesis of autoimmune exocrinopathy, we profiled a mouse model of spontaneous, autoimmune exocrinopathy that possess key characteristics of peripheral neuropathy experienced by SS patients. Mice deficient in the autoimmune regulator (Aire) gene developed spontaneous, CD4+ T cell-mediated exocrinopathy and aqueous-deficient dry eye that were associated with loss of nerves innervating the cornea and lacrimal gland. Changes in innervation and tear secretion were accompanied by increased proliferation of corneal epithelial basal cells, limbal expansion of KRT19-positive progenitor cells, increased vascularization of the peripheral cornea and reduced nerve function in the lacrimal gland. In addition, we found extensive loss of MIST1+ secretory acinar cells in the Aire -/- lacrimal gland suggesting that acinar cells are a primary target of the disease, Finally, topical application of ophthalmic steroid effectively restored corneal innervation in Aire -/- mice thereby functionally linking nerve loss with local inflammation in the aqueous-deficient dry eye. These data provide important insight regarding the relationship between chronic inflammation and neuropathic changes in autoimmune-mediated dry eye. Peripheral neuropathies characteristic of SS appear to be tightly linked with the underlying immunopathological mechanism and Aire -/- mice provide an excellent tool to explore the interplay between SS-associated immunopathology and peripheral neuropathy.


Subject(s)
Cornea/pathology , Lacrimal Apparatus/pathology , Sjogren's Syndrome/pathology , Transcription Factors/genetics , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cornea/cytology , Cornea/drug effects , Cornea/metabolism , Disease Models, Animal , Female , Humans , Lacrimal Apparatus/metabolism , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Neovascularization, Physiologic , Neurites/drug effects , Neurites/metabolism , Prednisolone/pharmacology , Prednisolone/therapeutic use , Sjogren's Syndrome/drug therapy , Sjogren's Syndrome/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Tears/metabolism , Transcription Factors/deficiency , AIRE Protein
19.
Elife ; 62017 06 17.
Article in English | MEDLINE | ID: mdl-28623666

ABSTRACT

Acinar cells play an essential role in the secretory function of exocrine organs. Despite this requirement, how acinar cells are generated during organogenesis is unclear. Using the acini-ductal network of the developing human and murine salivary gland, we demonstrate an unexpected role for SOX2 and parasympathetic nerves in generating the acinar lineage that has broad implications for epithelial morphogenesis. Despite SOX2 being expressed by progenitors that give rise to both acinar and duct cells, genetic ablation of SOX2 results in a failure to establish acini but not ducts. Furthermore, we show that SOX2 targets acinar-specific genes and is essential for the survival of acinar but not ductal cells. Finally, we illustrate an unexpected and novel role for peripheral nerves in the creation of acini throughout development via regulation of SOX2. Thus, SOX2 is a master regulator of the acinar cell lineage essential to the establishment of a functional organ.


Subject(s)
Acinar Cells/physiology , Cell Differentiation , Organogenesis , SOXB1 Transcription Factors/metabolism , Salivary Glands/cytology , Salivary Glands/embryology , Animals , Gene Knockout Techniques , Humans , Mice
20.
Sci Rep ; 7(1): 3484, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615711

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) play crucial roles in maintaining tissue homeostasis during physiological turnovers and injuries. Very little is known about the phenotype, distribution and molecular nature of MSCs in freshly isolated human salivary glands (SGs) as most reports have focused on the analysis of cultured MSCs. Our results demonstrate that the cell adhesion molecule CD34 was widely expressed by the MSCs of human major SGs, namely parotid (PAG), sublingual (SLG) and submandibular (SMG) glands. Further, gene expression analysis of CD34+ cells derived from fetal SMGs showed significant upregulation of genes involved in cellular adhesion, proliferation, branching, extracellular matrix remodeling and organ development. Moreover, CD34+ SMG cells exhibited elevated expression of genes encoding extracellular matrix, basement membrane proteins, and members of ERK, FGF and PDGF signaling pathways, which play key roles in glandular development, branching and homeostasis. In vitro CD34+ cell derived SG-MSCs revealed multilineage differentiation potential. Intraglandular transplantation of cultured MSCs in immunodeficient mice led to their engraftment in the injected and uninjected contralateral and ipsilateral glands. Engrafted cells could be localized to the stroma surrounding acini and ducts. In summary, our data show that CD34+ derived SG-MSCs could be a promising cell source for adoptive cell-based SG therapies, and bioengineering of artificial SGs.


Subject(s)
Antigens, CD34/metabolism , Mesenchymal Stem Cells/metabolism , Parotid Gland/metabolism , Sublingual Gland/metabolism , Submandibular Gland/metabolism , Adult , Animals , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Mesenchymal Stem Cell Transplantation , Mice , Middle Aged , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL