Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maxime M. Mahe is active.

Publication


Featured researches published by Maxime M. Mahe.


Nature Medicine | 2014

An in vivo model of human small intestine using pluripotent stem cells

Carey L. Watson; Maxime M. Mahe; Jorge O. Múnera; Jonathan C. Howell; Nambirajan Sundaram; Holly M. Poling; Jamie I. Schweitzer; Jefferson Vallance; Christopher N. Mayhew; Ying Sun; Gregory A. Grabowski; Stacy R. Finkbeiner; Jason R. Spence; Noah F. Shroyer; James M. Wells; Michael A. Helmrath

Differentiation of human pluripotent stem cells (hPSCs) into organ-specific subtypes offers an exciting avenue for the study of embryonic development and disease processes, for pharmacologic studies and as a potential resource for therapeutic transplant. To date, limited in vivo models exist for human intestine, all of which are dependent upon primary epithelial cultures or digested tissue from surgical biopsies that include mesenchymal cells transplanted on biodegradable scaffolds. Here, we generated human intestinal organoids (HIOs) produced in vitro from human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) that can engraft in vivo. These HIOs form mature human intestinal epithelium with intestinal stem cells contributing to the crypt-villus architecture and a laminated human mesenchyme, both supported by mouse vasculature ingrowth. In vivo transplantation resulted in marked expansion and maturation of the epithelium and mesenchyme, as demonstrated by differentiated intestinal cell lineages (enterocytes, goblet cells, Paneth cells, tuft cells and enteroendocrine cells), presence of functional brush-border enzymes (lactase, sucrase-isomaltase and dipeptidyl peptidase 4) and visible subepithelial and smooth muscle layers when compared with HIOs in vitro. Transplanted intestinal tissues demonstrated digestive functions as shown by permeability and peptide uptake studies. Furthermore, transplanted HIO-derived tissue was responsive to systemic signals from the host mouse following ileocecal resection, suggesting a role for circulating factors in the intestinal adaptive response. This model of the human small intestine may pave the way for studies of intestinal physiology, disease and translational studies.


Gastroenterology | 2013

Isolation and Characterization of Intestinal Stem Cells Based on Surface Marker Combinations and Colony-Formation Assay

Fengchao Wang; David Scoville; Xi C. He; Maxime M. Mahe; Andrew C. Box; John M. Perry; Nicholas R. Smith; Nan Ye Lei; Paige S. Davies; Megan K. Fuller; Jeffrey S. Haug; Melainia McClain; Adam D. Gracz; Sheng Ding; Matthias Stelzner; James C.Y. Dunn; Scott T. Magness; Melissa H. Wong; Martin G. Martin; Michael A. Helmrath; Linheng Li

BACKGROUND & AIMS Identification of intestinal stem cells (ISCs) has relied heavily on the use of transgenic reporters in mice, but this approach is limited by mosaic expression patterns and difficult to directly apply to human tissues. We sought to identify reliable surface markers of ISCs and establish a robust functional assay to characterize ISCs from mouse and human tissues. METHODS We used immunohistochemistry, real-time reverse-transcription polymerase chain reaction, and fluorescence-activated cell sorting (FACS) to analyze intestinal epithelial cells isolated from mouse and human intestinal tissues. We compared different combinations of surface markers among ISCs isolated based on expression of Lgr5-green fluorescent protein. We developed a culture protocol to facilitate the identification of functional ISCs from mice and then tested the assay with human intestinal crypts and putative ISCs. RESULTS CD44(+)CD24(lo)CD166(+) cells, isolated by FACS from mouse small intestine and colon, expressed high levels of stem cell-associated genes. Transit-amplifying cells and progenitor cells were then excluded based on expression of GRP78 or c-Kit. CD44(+)CD24(lo)CD166(+) GRP78(lo/-) putative stem cells from mouse small intestine included Lgr5-GFP(hi) and Lgr5-GFP(med/lo) cells. Incubation of these cells with the GSK inhibitor CHIR99021 and the E-cadherin stabilizer Thiazovivin resulted in colony formation by 25% to 30% of single-sorted ISCs. CONCLUSIONS We developed a culture protocol to identify putative ISCs from mouse and human tissues based on cell surface markers. CD44(+)CD24(lo)CD166(+), GRP78(lo/-), and c-Kit(-) facilitated identification of putative stem cells from the mouse small intestine and colon, respectively. CD44(+)CD24(-/lo)CD166(+) also identified putative human ISCs. These findings will facilitate functional studies of mouse and human ISCs.


Current protocols in mouse biology | 2013

Establishment of Gastrointestinal Epithelial Organoids

Maxime M. Mahe; Eitaro Aihara; Michael Schumacher; Yana Zavros; Marshall H. Montrose; Michael A. Helmrath; Toshiro Sato; Noah F. Shroyer

The intestinal epithelium constitutes a system of constant and rapid renewal triggered by proliferation of intestinal stem cells (ISCs), and is an ideal system for studying cell proliferation, migration, and differentiation. Primary cell cultures have proven to be promising for unraveling the mechanisms involved in epithelium homeostasis. In 2009, Sato et al. established a long‐term primary culture to generate epithelial organoids (enteroids) with crypt‐ and villus‐like epithelial domains representing the complete census of progenitors and differentiated cells. Similarly, isolated ISCs expressing Lgr5 (leucine‐rich repeat‐containing G protein–coupled receptor) can generate enteroids. Here, we describe methods to establish gastric, small intestinal, and colonic epithelial organoids and generate Lgr5+ve single cell–derived epithelial organoids. We also describe the imaging techniques used to characterize those organoids. This in vitro model constitutes a powerful tool for studying stem cell biology and intestinal epithelial cell physiology throughout the digestive tract. Curr. Protoc. Mouse Biol. 3:217‐240


Nature Medicine | 2017

Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system

Michael Workman; Maxime M. Mahe; Stephen L. Trisno; Holly M. Poling; Carey Watson; Nambirajan Sundaram; Ching Fang Chang; Jacqueline V. Schiesser; Philippe Aubert; Edouard G. Stanley; Andrew G. Elefanty; Yuichiro Miyaoka; Mohammad A. Mandegar; Bruce R. Conklin; Michel Neunlist; Samantha A. Brugmann; Michael A. Helmrath; James M. Wells

The enteric nervous system (ENS) of the gastrointestinal tract controls many diverse functions, including motility and epithelial permeability. Perturbations in ENS development or function are common, yet there is no human model for studying ENS-intestinal biology and disease. We used a tissue-engineering approach with embryonic and induced pluripotent stem cells (PSCs) to generate human intestinal tissue containing a functional ENS. We recapitulated normal intestinal ENS development by combining human-PSC-derived neural crest cells (NCCs) and developing human intestinal organoids (HIOs). NCCs recombined with HIOs in vitro migrated into the mesenchyme, differentiated into neurons and glial cells and showed neuronal activity, as measured by rhythmic waves of calcium transients. ENS-containing HIOs grown in vivo formed neuroglial structures similar to a myenteric and submucosal plexus, had functional interstitial cells of Cajal and had an electromechanical coupling that regulated waves of propagating contraction. Finally, we used this system to investigate the cellular and molecular basis for Hirschsprungs disease caused by a mutation in the gene PHOX2B. This is, to the best of our knowledge, the first demonstration of human-PSC-derived intestinal tissue with a functional ENS and how this system can be used to study motility disorders of the human gastrointestinal tract.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Enteric glia promote intestinal mucosal healing via activation of focal adhesion kinase and release of proEGF

Laurianne Van Landeghem; Julien Chevalier; Maxime M. Mahe; Thilo Wedel; Petri Urvil; Pascal Derkinderen; Tor C. Savidge; Michel Neunlist

Wound healing of the gastrointestinal mucosa is essential for the maintenance of gut homeostasis and integrity. Enteric glial cells play a major role in regulating intestinal barrier function, but their role in mucosal barrier repair remains unknown. The impact of conditional ablation of enteric glia on dextran sodium sulfate (DSS)-induced mucosal damage and on healing of diclofenac-induced mucosal ulcerations was evaluated in vivo in GFAP-HSVtk transgenic mice. A mechanically induced model of intestinal wound healing was developed to study glial-induced epithelial restitution. Glial-epithelial signaling mechanisms were analyzed by using pharmacological inhibitors, neutralizing antibodies, and genetically engineered intestinal epithelial cells. Enteric glial cells were shown to be abundant in the gut mucosa, where they associate closely with intestinal epithelial cells as a distinct cell population from myofibroblasts. Conditional ablation of enteric glia worsened mucosal damage after DSS treatment and significantly delayed mucosal wound healing following diclofenac-induced small intestinal enteropathy in transgenic mice. Enteric glial cells enhanced epithelial restitution and cell spreading in vitro. These enhanced repair processes were reproduced by use of glial-conditioned media, and soluble proEGF was identified as a secreted glial mediator leading to consecutive activation of epidermal growth factor receptor and focal adhesion kinase signaling pathways in intestinal epithelial cells. Our study shows that enteric glia represent a functionally important cellular component of the intestinal epithelial barrier microenvironment and that the disruption of this cellular network attenuates the mucosal healing process.


Stem cell reports | 2015

Transcriptome-wide Analysis Reveals Hallmarks of Human Intestine Development and Maturation In Vitro and In Vivo.

Stacy R. Finkbeiner; David R. Hill; Christopher H. Altheim; Priya H. Dedhia; Matthew Taylor; Yu Hwai Tsai; Alana M. Chin; Maxime M. Mahe; Carey L. Watson; Jennifer J. Freeman; Roy Nattiv; Matthew Thomson; Ophir D. Klein; Noah F. Shroyer; Michael A. Helmrath; Daniel H. Teitelbaum; Peter J. Dempsey; Jason R. Spence

Summary Human intestinal organoids (HIOs) are a tissue culture model in which small intestine-like tissue is generated from pluripotent stem cells. By carrying out unsupervised hierarchical clustering of RNA-sequencing data, we demonstrate that HIOs most closely resemble human fetal intestine. We observed that genes involved in digestive tract development are enriched in both fetal intestine and HIOs compared to adult tissue, whereas genes related to digestive function and Paneth cell host defense are expressed at higher levels in adult intestine. Our study also revealed that the intestinal stem cell marker OLFM4 is expressed at very low levels in fetal intestine and in HIOs, but is robust in adult crypts. We validated our findings using in vivo transplantation to show that HIOs become more adult-like after transplantation. Our study emphasizes important maturation events that occur in the intestine during human development and demonstrates that HIOs can be used to model fetal-to-adult maturation.


PLOS Pathogens | 2015

CD44 plays a functional role in Helicobacter pylori-induced epithelial cell proliferation.

Nina Bertaux-Skeirik; Rui Feng; Michael Schumacher; Jing Li; Maxime M. Mahe; Amy C. Engevik; Jose Javier; Richard M. Peek; Karen M. Ottemann; Véronique Orian-Rousseau; Gregory P. Boivin; Michael A. Helmrath; Yana Zavros

The cytotoxin-associated gene (Cag) pathogenicity island is a strain-specific constituent of Helicobacter pylori (H. pylori) that augments cancer risk. CagA translocates into the cytoplasm where it stimulates cell signaling through the interaction with tyrosine kinase c-Met receptor, leading cellular proliferation. Identified as a potential gastric stem cell marker, cluster-of-differentiation (CD) CD44 also acts as a co-receptor for c-Met, but whether it plays a functional role in H. pylori-induced epithelial proliferation is unknown. We tested the hypothesis that CD44 plays a functional role in H. pylori-induced epithelial cell proliferation. To assay changes in gastric epithelial cell proliferation in relation to the direct interaction with H. pylori, human- and mouse-derived gastric organoids were infected with the G27 H. pylori strain or a mutant G27 strain bearing cagA deletion (∆CagA::cat). Epithelial proliferation was quantified by EdU immunostaining. Phosphorylation of c-Met was analyzed by immunoprecipitation followed by Western blot analysis for expression of CD44 and CagA. H. pylori infection of both mouse- and human-derived gastric organoids induced epithelial proliferation that correlated with c-Met phosphorylation. CagA and CD44 co-immunoprecipitated with phosphorylated c-Met. The formation of this complex did not occur in organoids infected with ∆CagA::cat. Epithelial proliferation in response to H. pylori infection was lost in infected organoids derived from CD44-deficient mouse stomachs. Human-derived fundic gastric organoids exhibited an induction in proliferation when infected with H. pylorithat was not seen in organoids pre-treated with a peptide inhibitor specific to CD44. In the well-established Mongolian gerbil model of gastric cancer, animals treated with CD44 peptide inhibitor Pep1, resulted in the inhibition of H. pylori-induced proliferation and associated atrophic gastritis. The current study reports a unique approach to study H. pylori interaction with the human gastric epithelium. Here, we show that CD44 plays a functional role in H. pylori-induced epithelial cell proliferation.


Journal of Visualized Experiments | 2015

Establishment of Human Epithelial Enteroids and Colonoids from Whole Tissue and Biopsy

Maxime M. Mahe; Nambirajan Sundaram; Carey L. Watson; Noah F. Shroyer; Michael A. Helmrath

The epithelium of the gastrointestinal tract is constantly renewed as it turns over. This process is triggered by the proliferation of intestinal stem cells (ISCs) and progeny that progressively migrate and differentiate toward the tip of the villi. These processes, essential for gastrointestinal homeostasis, have been extensively studied using multiple approaches. Ex vivo technologies, especially primary cell cultures have proven to be promising for understanding intestinal epithelial functions. A long-term primary culture system for mouse intestinal crypts has been established to generate 3-dimensional epithelial organoids. These epithelial structures contain crypt- and villus-like domains reminiscent of normal gut epithelium. Commonly, termed “enteroids” when derived from small intestine and “colonoids” when derived from colon, they are different from organoids that also contain mesenchyme tissue. Additionally, these enteroids/colonoids continuously produce all cell types found normally within the intestinal epithelium. This in vitro organ-like culture system is rapidly becoming the new gold standard for investigation of intestinal stem cell biology and epithelial cell physiology. This technology has been recently transferred to the study of human gut. The establishment of human derived epithelial enteroids and colonoids from small intestine and colon has been possible through the utilization of specific culture media that allow their growth and maintenance over time. Here, we describe a method to establish a small intestinal and colon crypt-derived system from human whole tissue or biopsies. We emphasize the culture modalities that are essential for the successful growth and maintenance of human enteroids and colonoids.


Cellular and molecular gastroenterology and hepatology | 2016

The Development of Spasmolytic Polypeptide/TFF2-Expressing Metaplasia (SPEM) During Gastric Repair Is Absent in the Aged Stomach

Amy C. Engevik; Rui Feng; Eun-Young Choi; Shana White; Nina Bertaux-Skeirik; Jing Li; Maxime M. Mahe; Eitaro Aihara; Li Yang; Betsy DiPasquale; Sunghee Oh; Kristen A. Engevik; Andrew S. Giraud; Marshall H. Montrose; Mario Medvedovic; Michael A. Helmrath; James R. Goldenring; Yana Zavros

Background & Aims During aging, physiological changes in the stomach result in more tenuous gastric tissue that is less capable of repairing injury, leading to increased susceptibility to chronic ulceration. Spasmolytic polypeptide/trefoil factor 2–expressing metaplasia (SPEM) is known to emerge after parietal cell loss and during Helicobacter pylori infection, however, its role in gastric ulcer repair is unknown. Therefore, we sought to investigate if SPEM plays a role in epithelial regeneration. Methods Acetic acid ulcers were induced in young (2–3 mo) and aged (18–24 mo) C57BL/6 mice to determine the quality of ulcer repair with advancing age. Yellow chameleon 3.0 mice were used to generate yellow fluorescent protein–expressing organoids for transplantation. Yellow fluorescent protein–positive gastric organoids were transplanted into the submucosa and lumen of the stomach immediately after ulcer induction. Gastric tissue was collected and analyzed to determine the engraftment of organoid-derived cells within the regenerating epithelium. Results Wound healing in young mice coincided with the emergence of SPEM within the ulcerated region, a response that was absent in the aged stomach. Although aged mice showed less metaplasia surrounding the ulcerated tissue, organoid-transplanted aged mice showed regenerated gastric glands containing organoid-derived cells. Organoid transplantation in the aged mice led to the emergence of SPEM and gastric regeneration. Conclusions These data show the development of SPEM during gastric repair in response to injury that is absent in the aged stomach. In addition, gastric organoids in an injury/transplantation mouse model promoted gastric regeneration.


Cell Reports | 2017

Canonical Wnt Signaling Ameliorates Aging of Intestinal Stem Cells

Kodandaramireddy Nalapareddy; Kalpana Nattamai; R. Kumar; Rebekah Karns; Kathryn A. Wikenheiser-Brokamp; Leesa Sampson; Maxime M. Mahe; Nambirajan Sundaram; Mary-Beth Yacyshyn; Bruce R. Yacyshyn; Michael A. Helmrath; Yi Zheng; Hartmut Geiger

SUMMARY Although intestinal homeostasis is maintained by intestinal stem cells (ISCs), regeneration is impaired upon aging. Here, we first uncover changes in intestinal architecture, cell number, and cell composition upon aging. Second, we identify a decline in the regenerative capacity of ISCs upon aging because of a decline in canonical Wnt signaling in ISCs. Changes in expression of Wnts are found in stem cells themselves and in their niche, including Paneth cells and mesenchyme. Third, reactivating canonical Wnt signaling enhances the function of both murine and human ISCs and, thus, ameliorates aging-associated phenotypes of ISCs in an organoid assay. Our data demonstrate a role for impaired Wnt signaling in physiological aging of ISCs and further identify potential therapeutic avenues to improve ISC regenerative potential upon aging.

Collaboration


Dive into the Maxime M. Mahe's collaboration.

Top Co-Authors

Avatar

Michael A. Helmrath

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nambirajan Sundaram

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Noah F. Shroyer

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yana Zavros

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Holly M. Poling

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

James M. Wells

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar

Amy C. Engevik

University of Cincinnati Academic Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eitaro Aihara

University of Cincinnati

View shared research outputs
Researchain Logo
Decentralizing Knowledge