Dennis L. Kasper’s research while affiliated with Harvard Medical School and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (450)


Binding properties of anti-huRGMb antibodies.
RGMb mAbs 2C11 and 5C10 inhibit RGMb interaction with PD-L2.
RGMb mAbs 2C11 and 5C10 differentially block RGMb interactions with BMPs and Neo1.
Differential binding of mAbs 2C11 and 5C10 to cell-surface RGMb.
Anticipated epitopes of RGMb mAbs 2C11 and 5C10 and PD-L2-binding interface in relation to BMP2-, BMP4-, Neo1-, and PD-L2-binding regions of RGMb.

+2

Targeting RGMb interactions: Discovery and preclinical characterization of potent anti-RGMb antibodies blocking multiple ligand bindings
  • Article
  • Full-text available

November 2024

·

13 Reads

Maria Meira

·

Aurore Frey

·

Neila Chekkat

·

[...]

·

Abdijapar Shamshiev

Therapeutic efficacy with durable responses has been demonstrated with several antibody drugs that block key immune checkpoint receptors, including PD-1, PD-L1, and CTLA-4. Despite the success of these drugs, a substantial proportion of patients do not benefit. Targeting multiple inhibitory pathways simultaneously to augment anti-tumor immunity has proven to be a promising approach. The emergence of Repulsive Guidance Molecule b (RGMb), a ligand for PD-L2, as a novel co-inhibitory pathway in T cells, together with its regulation by the gut microbiome, encouraged the discovery and development of fully human anti-RGMb antibodies. Here, we describe phage display-derived monoclonal antibodies (mAbs) 2C11 and 5C10 that bind human RGMb with high affinities of 1.4 nM and 0.72 nM, respectively. Both mAbs 2C11 and 5C10 potently inhibited RGMb interaction with PD-L2. MAb 2C11 effectively inhibited RGMb interaction with bone morphogenetic proteins 2 and 4 (BMP2–4), while leaving RGMb interaction with Neogenin 1 (Neo1) unaffected. Conversely, mAb 5C10 disrupted RGMb interaction with Neo1 while maintaining RGMb binding to BMP2–4. These findings map the 2C11 epitope at the membrane-distal N-terminal region of RGMb, which coincides with both PD-L2- and BMP2–4-binding sites. The PD-L2 binding interface is likely positioned between RGMb’s N-terminal BMP-binding and C-terminal Neo1-binding regions. The in vivo activity of mAb 2C11 in combination with anti-PD-1 or anti-PD-L1 was tested in MC38 and B16-OVA cancer models and demonstrated synergistic effects by significantly enhancing anti-tumor responses. These properties make mAb 2C11 a promising candidate for therapeutic use to overcome immune checkpoint inhibitor resistances, warranting further exploration in clinical settings.

Download

Figure 4. Evidence of microbial production of N-acylated histamines. Concentrations of (A) histamine-
The microbiome diversifies N-acyl lipid pools - including short-chain fatty acid-derived compounds

November 2024

·

107 Reads

N-acyl lipids are important mediators of several biological processes including immune function and stress response. To enhance the detection of N-acyl lipids with untargeted mass spectrometry-based metabolomics, we created a reference spectral library retrieving N-acyl lipid patterns from 2,700 public datasets, identifying 851 N-acyl lipids that were detected 356,542 times. 777 are not documented in lipid structural databases, with 18% of these derived from short-chain fatty acids and found in the digestive tract and other organs. Their levels varied with diet, microbial colonization, and in people living with diabetes. We used the library to link microbial N-acyl lipids, including histamine and polyamine conjugates, to HIV status and cognitive impairment. This resource will enhance the annotation of these compounds in future studies to further the understanding of their roles in health and disease and highlight the value of large-scale untargeted metabolomics data for metabolite discovery.


Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production

May 2024

·

31 Reads

·

2 Citations

Cellular and Molecular Gastroenterology and Hepatology

Background & Aims Gut bacterial sphingolipids, primarily produced by Bacteroidetes, have dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells. Patients with inflammatory bowel disease display altered sphingolipids profiles in fecal samples. However, how bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remains unclear. Methods We used dextran sodium sulfate (DSS)-induced colitis in mice monocolonized with Bacteroides fragilis strains expressing or lacking sphingolipids to assess the influence of bacterial sphingolipids on intestinal inflammation using transcriptional, protein, and cellular analyses. Colonic explant and organoid were used to study the function of bacterial sphingolipids. Host mucosal immune cells and cytokines were profiled and characterized using flow cytometry, enzyme-linked immunosorbent assay, and Western blot, and cytokine function in vivo was investigated by monoclonal antibody injection. Results B fragilis sphingolipids exacerbated intestinal inflammation. Mice monocolonized with B fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin (IL)-22 by ILC3. Mice colonized with B fragilis lacking sphingolipids following DSS treatment showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production. Protection against DSS colitis associated with B fragilis lacking sphingolipids was reversed on IL22 blockade. Furthermore, bacterial sphingolipids restricted epithelial IL18 production following DSS treatment and interfered with IL22 production by a subset of ILC3 cells expressing both IL18R and major histocompatibility complex class II. Conclusions B fragilis–derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL18 expression and concomitantly suppressing the production of IL22 by ILC3 cells.


Top-down Mass Spectrometric Analysis of a Lipooligosaccharide from the Human Commensal Bacteroides fragilis

April 2024

·

25 Reads

·

1 Citation

Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are ubiquitous structures found on the outer membrane of gram-negative bacteria. Bacteroides fragilis , is a gram-negative anaerobe commonly inhabiting the human colon. The LOS of this organism is known to trigger a type I interferon response in dendritic cells. However, detailed structural analysis of this LOS has been largely elusive. Using top-down mass spectrometry, we have unraveled the comprehensive fine structure of B. fragilis LOS. Our analysis reveals that this LOS has a poly-galactose-rhamnose-KDO-lipid A architecture, which can be modified by hexuronic acid and ethanolamine via phosphodiester linkages. The lipid moiety typically includes three to five acyl chains of varying length on a glucosamine disaccharide. This investigation lays the groundwork for deeper immunological exploration of B. fragilis LOS and underscores the efficacy of top-down mass spectrometry in characterizing intact LOS/LPS structures and their modifications.




BACTERIAL SPHINGOLIPIDS EXACERBATE COLITIS BY INHIBITING ILC3-DERIVED IL-22 PRODUCTION

January 2024

·

12 Reads

Inflammatory Bowel Diseases

Commensal bacteria of the Bacteroidetes phylum are the primary producers of sphingolipids in the gut lumen. These lipids serve dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells (iNKT). Sphingolipid composition is significantly altered in fecal samples of patients with inflammatory bowel disease (IBD). However, the specific mechanisms by which bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remain unclear. In this study, we investigated the impact of bacterial sphingolipids on intestinal inflammation by mono-colonizing mice with Bacteroides fragilis strains that either express or lack sphingolipids during DSS-induced colitis. We discovered that B. fragilis sphingolipids exacerbate intestinal inflammation. Mice mono-colonized with B. fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin-22 (IL-22) by innate lymphoid cell type 3 (ILC3). Consistent with the inhibitory effect of sphingolipids on IL-22 production, mice colonized with B. fragilis lacking sphingolipids showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production following DSS treatment compared to those colonized with B. fragilis producing sphingolipids. Additionally, colitis severity in mice colonized with B. fragilis lacking sphingolipids was exacerbated upon IL-22 blockade. Furthermore, our study reveals that bacterial sphingolipids restrict epithelial IL-18 production following DSS treatment and interfere with IL-22 production by a subset of ILC3 cells expressing both the interleukin-18 receptor (IL-18R) and major histocompatibility complex class II (MHC II). These findings indicate that B. fragilis-derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL-18 expression, resulting in compromised production of IL-22 by ILC3 cells. Work Model



TNFR superfamily costimulatory molecules.
Microbiota-dependent regulation of costimulatory and coinhibitory pathways via innate immune sensors and implications for immunotherapy

September 2023

·

96 Reads

·

8 Citations

Experimental and Molecular Medicine

Our bodies are inhabited by trillions of microorganisms. The host immune system constantly interacts with the microbiota in barrier organs, including the intestines. Over decades, numerous studies have shown that our mucosal immune system is dynamically shaped by a variety of microbiota-derived signals. Elucidating the mediators of these interactions is an important step for understanding how the microbiota is linked to mucosal immune homeostasis and gut-associated diseases. Interestingly, the efficacy of cancer immunotherapies that manipulate costimulatory and coinhibitory pathways has been correlated with the gut microbiota. Moreover, adverse effects of these therapies in the gut are linked to dysregulation of the intestinal immune system. These findings suggest that costimulatory pathways in the immune system might serve as a bridge between the host immune system and the gut microbiota. Here, we review mechanisms by which commensal microorganisms signal immune cells and their potential impact on costimulation. We highlight how costimulatory pathways modulate the mucosal immune system through not only classical antigen-presenting cells but also innate lymphocytes, which are highly enriched in barrier organs. Finally, we discuss the adverse effects of immune checkpoint inhibitors in the gut and the possible relationship with the gut microbiota.


Bacterial Sphingolipids Exacerbate Colitis by Inhibiting ILC3-derived IL-22 Production

September 2023

·

56 Reads

Commensal bacteria of the Bacteroidetes phylum are the primary producers of sphingolipids in the gut lumen. These lipids serve dual roles as bacterial virulence factors and regulators of the host mucosal immune system, including regulatory T cells and invariant natural killer T cells (iNKT). Sphingolipid composition is significantly altered in fecal samples of patients with inflammatory bowel disease (IBD). However, the specific mechanisms by which bacterial sphingolipids modulate mucosal homeostasis and regulate intestinal inflammation remain unclear. In this study, we investigated the impact of bacterial sphingolipids on intestinal inflammation by mono-colonizing mice with Bacteroides fragilis strains that either express or lack sphingolipids during DSS-induced colitis. We discovered that B. fragilis sphingolipids exacerbate intestinal inflammation. Mice mono-colonized with B. fragilis lacking sphingolipids exhibited less severe DSS-induced colitis. This amelioration of colitis was associated with increased production of interleukin-22 (IL-22) by innate lymphoid cell type 3 (ILC3). Consistent with the inhibitory effect of sphingolipids on IL-22 production, mice colonized with B. fragilis lacking sphingolipids showed enhanced epithelial STAT3 activity, intestinal cell proliferation, and antimicrobial peptide production following DSS treatment compared to those colonized with B. fragilis producing sphingolipids. Additionally, colitis severity in mice colonized with B. fragilis lacking sphingolipids was exacerbated upon IL-22 blockade. Furthermore, our study reveals that bacterial sphingolipids restrict epithelial IL-18 production following DSS treatment and interfere with IL-22 production by a subset of ILC3 cells expressing both the interleukin-18 receptor (IL-18R) and major histocompatibility complex class II (MHC II). These findings indicate that B. fragilis -derived sphingolipids exacerbate mucosal inflammation by impeding epithelial IL-18 expression, resulting in compromised production of IL-22 by ILC3 cells. Highlights B. fragilis -derived sphingolipids exacerbate DSS-induced colitis in mono-colonized C57BL/6 mice. B. fragilis -derived sphingolipids constrain ILC3-derived IL-22, leading to reduced colonic epithelial cell proliferation and compromised barrier function. B. fragilis -derived sphingolipids restrict epithelial NLRC4 inflammasome activation and IL-18 secretion. B. fragilis -derived sphingolipids modulate IL-22 production by IL18R ⁺ MHC II ⁺ ILC3s.


Citations (69)


... Lately, more researchers have concentrated on the connection between bile acids and cancer development (Cong et al., 2024;Zhu et al., 2023;Riscal et al., 2024). Cholesterol's primary metabolites, bile acids, are produced in the liver and traveled to the intestine via the enterohepatic circulation. ...

Reference:

Hyodeoxycholic acid inhibits colorectal cancer proliferation through the FXR/EREG/EGFR axis
Bile acids modified by the intestinal microbiota promote colorectal cancer growth by suppressing CD8+ T cell effector functions
  • Citing Article
  • March 2024

Immunity

... An influential environmental factor that shapes physiology 3,4 and which has evolved in a bidirectional and mutualistic relationship with its host 5,6 is the commensal microbiota (Box 2). This densely colonized microbial ecosystem is locally sensed by host tissues and communicates to remote organs and affects their functions, such as via the gut-liver axis 7,8 . ...

Gut complement induced by the microbiota combats pathogens and spares commensals
  • Citing Article
  • January 2024

Cell

... Recently, there is increasing evidence suggesting that the effectiveness of cancer immunotherapies has been associated with the gut microbiota (Park, Gazzaniga, Kasper, & Sharpe, 2023). Changes in the composition of the microbiome can disrupt immune regulation, leading to chronic inflammation and contributing to tumor development. ...

Microbiota-dependent regulation of costimulatory and coinhibitory pathways via innate immune sensors and implications for immunotherapy

Experimental and Molecular Medicine

... In a healthy state, the gut microbial community maintains dynamic balance and exhibits high taxonomic diversity, rich microbial gene abundance, and a stable functional core of the microbiome. The integration of its derived nutritional signals into the first-line defense system at the mucosal interface results in the production of a wide range of beneficial metabolites, which participate in stimulating the expression of multiple HDPs (Wu et al. 2020;Song et al. 2023). ...

Gut microbial fatty acid isomerization modulates intraepithelial T cells

Nature

... In addition to shaping the inflammatory microenvironment, GM can also influence the immune barrier. Sharpe and colleagues found that a specific gut bacterium, Coprobacillus cateniformis, enhances the efficacy of PD-1 checkpoint blockade therapy by downregulating the expression and activity of immune molecules PD-L2 and repulsive guidance molecule b [36]. Balancing and optimizing the GM could have a significant impact on controlling the development of tumors. ...

Publisher Correction: Targeting PD-L2–RGMb overcomes microbiome-related immunotherapy resistance

Nature

... Previous studies have identified two distinct pathways of bacterial sphingolipid synthesis from 3-KS (26,37). As illustrated in Fig. 1A, one pathway involves 3-ketosphin ganine reductase (3-KDSR), catalyzing the conversion of 3-KS to sphinganine, followed by the reaction catalyzed by ceramide synthase (CerS), which converts this precursor into dihydroceramides (38). ...

Functional and metagenomic level diversities of human gut symbiont-derived glycolipids
  • Citing Preprint
  • May 2023

... Emerging evidence from numerous studies highlights a connection between the symbiotic microbiome and cancer, 21 with compelling data suggesting that the gastrointestinal microbiome plays a critical role in modulating responses to cancer immunotherapy. [22][23][24][25][26][27] Recent research has focused increasing attention on the pivotal role of the gut microbiome in modulating immune responses, [28][29][30][31][32] particularly in the context of cancer therapies. Studies have demonstrated that specific microbial compositions can enhance the efficacy of immune checkpoint inhibitors and other forms of immunotherapy by influencing the host immune system. ...

Targeting PD-L2–RGMb overcomes microbiome-related immunotherapy resistance

Nature

... Although most bacteria can efficiently colonize in the humanized mice after FMT, current FMT experiments cannot fully simulate the gut microbiota of the donor (52)(53)(54)(55)(56). ...

Kinetic and mechanistic diversity of intestinal immune homeostasis characterized by rapid removal of gut bacteria

... Complement proteins have been detected in the gut lumen (54), and it has been suggested that they are activated and regulated by infiltrating cells; properdin, for example, is an alternative pathway complement regulatory protein produced by activated human neutrophils and secreted upon inflammatory cytokine signaling (55,56). A recent study from Dennis Kasper lab showed that complement C3 was synthesized by three key types of cells in a microbiome-dependent manner and secreted into the intestinal lumen; the group reported luminal C3 to be critical for protection against enteric pathogens in mice infected with either Citrobacter rodentium or enterohemorrhagic Escherichia coli (57). ...

Microbiome induced complement synthesized in the gut protects against enteric infections
  • Citing Preprint
  • February 2023

... We hypothesize that the decrease in both neuropeptides may be due to the loss of corneal nerve fibers resulting from corneal damage in dry eye patients, leading to reduced neuropeptide synthesis and subsequently decreased release of sP and cGRP. additionally, research has suggested a potential intrinsic connection and regulatory relationship between cGRP and goblet cell secretion in the intestinal system [22], while the deficiency of mucin secretion from conjunctival goblet cells is one of the main pathogenic factors in dry eye patients. this suggests a potential inherent connection and regulatory relationship between cGRP and secretion of tears. ...

Nociceptor neurons direct goblet cells via a CGRP-RAMP1 axis to drive mucus production and gut barrier protection
  • Citing Article
  • October 2022

Cell